WO1993007283A1 - Zusammensetzung für das einbringen von nukleinsäure-komplexen in höhere eukaryotische zellen - Google Patents

Zusammensetzung für das einbringen von nukleinsäure-komplexen in höhere eukaryotische zellen Download PDF

Info

Publication number
WO1993007283A1
WO1993007283A1 PCT/EP1992/002234 EP9202234W WO9307283A1 WO 1993007283 A1 WO1993007283 A1 WO 1993007283A1 EP 9202234 W EP9202234 W EP 9202234W WO 9307283 A1 WO9307283 A1 WO 9307283A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
gly
virus
nucleic acid
adenovirus
Prior art date
Application number
PCT/EP1992/002234
Other languages
German (de)
English (en)
French (fr)
Inventor
David Curiel
Ernst Wagner
Matt Cotten
Kurt Zatloukal
Christian Plank
Max L. Birnstiel
Bernd Oberhauser
Walter G. M. Schmidt
Original Assignee
Boehringer Ingelheim International Gmbh
Genentech Inc.
The University Of North Carolina
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to JP5506590A priority Critical patent/JPH10506001A/ja
Priority to RO94-00499A priority patent/RO117861B1/ro
Application filed by Boehringer Ingelheim International Gmbh, Genentech Inc., The University Of North Carolina filed Critical Boehringer Ingelheim International Gmbh
Priority to EP92920580A priority patent/EP0607206B1/de
Priority to CA002118816A priority patent/CA2118816C/en
Priority to AT92920580T priority patent/ATE215990T1/de
Priority to SK368-94A priority patent/SK281682B6/sk
Priority to BR9206559A priority patent/BR9206559A/pt
Priority to AU26526/92A priority patent/AU671084B2/en
Priority to ES92920580T priority patent/ES2173083T3/es
Priority to DE59209951T priority patent/DE59209951D1/de
Priority to PL92303106A priority patent/PL180304B1/pl
Priority to DK92920580T priority patent/DK0607206T3/da
Priority to KR1019940700967A priority patent/KR100241685B1/ko
Publication of WO1993007283A1 publication Critical patent/WO1993007283A1/de
Priority to NO19941154A priority patent/NO316744B1/no
Priority to FI941474A priority patent/FI941474A0/fi
Priority to BG98718A priority patent/BG62740B1/bg
Priority to HK98113354A priority patent/HK1013104A1/xx

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6901Conjugates being cells, cell fragments, viruses, ghosts, red blood cells or viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/775Apolipopeptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/79Transferrins, e.g. lactoferrins, ovotransferrins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10211Aviadenovirus, e.g. fowl adenovirus A
    • C12N2710/10251Methods of production or purification of viral material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10211Aviadenovirus, e.g. fowl adenovirus A
    • C12N2710/10261Methods of inactivation or attenuation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10322New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10351Methods of production or purification of viral material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10361Methods of inactivation or attenuation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10361Methods of inactivation or attenuation
    • C12N2710/10363Methods of inactivation or attenuation by chemical treatment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/11011Alpharetrovirus, e.g. avian leucosis virus
    • C12N2740/11022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/32011Picornaviridae
    • C12N2770/32711Rhinovirus
    • C12N2770/32722New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/32011Picornaviridae
    • C12N2770/32711Rhinovirus
    • C12N2770/32761Methods of inactivation or attenuation
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/14Heterocyclic carbon compound [i.e., O, S, N, Se, Te, as only ring hetero atom]
    • Y10T436/142222Hetero-O [e.g., ascorbic acid, etc.]
    • Y10T436/143333Saccharide [e.g., DNA, etc.]

Definitions

  • composition for the introduction of nucleic acid complexes into higher eukaryotic cells Composition for the introduction of nucleic acid complexes into higher eukaryotic cells
  • the invention relates to the introduction of nucleic acids into higher eukaryotic cells.
  • genetic diseases in which gene therapy is a promising approach are hemophilia, beta-thalassemia and Severe Combined Immune Deficiency (SCID), a syndrome caused by a genetic deficiency of the enzyme adenosine deaminase.
  • SCID Severe Combined Immune Deficiency
  • nucleic acid which codes for a secreted protein antigen or for a non-secreted protein antigen, by means of a vaccination to achieve a humoral or intracellular immunity.
  • Other examples of genetic defects where administration of nucleic acid encoding the defective gene e.g. Muscular dystrophy (dystrophin gene), cystic fibrosis can be administered in an individually tailored form
  • REPLACEMENT LEAF Certhelial fibrosis transmembrane conductance regulator gene
  • Hypercholesterolä ie DL receptor gene
  • Gene therapy is also a promising approach for the treatment of cancer, with so-called “cancer vaccines” being administered.
  • cancer vaccines To increase the immunogenicity of tumor cells, they are altered to either make them more antigenic or to cause them to produce certain immunomodulating substances, e.g. Cytokines, which then trigger an immune response.
  • the cells are transfected with DNA, which is essential for a cytokine, e.g. IL-2, IL-4, IFN-gamma, TNF- ⁇ . So far, gene transfer to autologous tumor cells has mainly been carried out using retroviral vectors.
  • retroviral systems for the transfer of genes into the cell (Wilson et al., 1990, Kasid et al., 1990).
  • retroviruses are problematic because, at least to a small extent, they involve the risk of side effects such as infection with the virus (due to recombination with endogenous viruses or contamination with helper viruses and possible subsequent mutation to a pathogenic form) or the development of cancer hides.
  • the stable transformation of the patient's somatic cells, as achieved with the aid of retroviruses is not always desirable because it does so the treatment, for example when side effects occur, can only be reversed with difficulty. It is also difficult with this form of therapy to obtain a sufficiently high titer to infect enough cells.
  • Nucleic acids as therapeutically active substances are also used to inhibit certain cell functions, e.g. Antisense RNAs and DNAs have proven to be effective agents for the selective inhibition of certain gene sequences. Their mode of action enables their use as therapeutic agents for blocking the expression of certain genes (such as deregulated oncogenes or viral genes) in vivo. It has already been shown that short antisense oligonucleotides can be imported into cells and exert their inhibitory effect there (Zamecnik et al., 1986), although their intracellular concentration, i.a. because of their limited uptake by the cell membrane due to the strong negative charge of the nucleic acids.
  • Another approach to selectively inhibit genes is to use ribozymes. Here too there is a need to ensure the highest possible concentration of active ribozymes in the cell, for which the transport into the cell is one of the limiting factors.
  • RNA decoys genes that have a protective effect against viruses.
  • protective genes e.g. transdominant mutations of genes that code for viral proteins or DNA molecules that code for so-called "RNA decoys”.
  • DNA transported into the cell is expressed by means of this system and that the inhibitory effect is not impaired by the transport system if nucleic acid having an inhibitory effect is used.
  • PCT application W091 / 17773 relates to a system for the transport of nucleic acids with a specific action for T cells.
  • This system makes use of cell surface proteins of the T cell lineage, e.g. CD4, the receptor used by the HIV virus.
  • the nucleic acid to be imported is complexed with a protein-polycation conjugate, the protein portion of which is a protein with the ability to bind to the T cell surface protein, e.g. CD4, bind and cells that express this surface protein brought into contact with the protein polycation / nucleic acid complexes obtained. It could be demonstrated that DNA transported into the cell is expressed in the cell with the aid of this system.
  • TZBLATT Common to these two inventions is the feature that they use specific cell functions to enable or facilitate the import of the nucleic acid into the cell.
  • the uptake mechanisms take place with the participation of factors which are referred to in the context of the present invention as "internalizing factors”. This includes factors that bind to the cell surface and are internalized, in the narrower or broader sense specific to the cell type, possibly with the participation of other factors (e.g. cell surface proteins). (In the case of the two inventions mentioned above, the
  • the internalization factor is conjugated with a substance of polycationic character which, due to its affinity for nucleic acids, creates a connection between the internalization factor and the nucleic acid.
  • a substance of polycationic character which, due to its affinity for nucleic acids, creates a connection between the internalization factor and the nucleic acid.
  • nucleic acid was chosen such that the internalizing factor polycation / nucleic acid complexes were largely electroneutral. Based on this observation, the methods for importing nucleic acids into higher ones were improved use eukaryotic cells internalizing factor-connecting factor / nucleic acid complexes.
  • nucleic acid-affine substance also increases the efficiency of the import system when another nucleic acid-affine substance is used as a connecting factor.
  • the complexes described by Wagner et al., 1991a which are taken up into higher eukaryotic cells by means of internalizing factor via endocytosis, contain nucleic acid, complexed with an internalizing factor-connecting factor conjuga.
  • the complexes contain one or more
  • nucleic acid-affine substances which may be identical to the compound factor, in a non-covalently bound form in such a way that the internalization and / or expression of the nucleic acid achieved by the conjugate is increased, which is primarily due to a condensing effect, but possibly also other mechanisms may be due.
  • chloroguin increases the pH in the lysosomes; on the basis of various experiments, it was found that other substances which, like chloroguin, have the ability to modulate the pH, such as monensin, ammonium chloride or methylamine, could not replace chloroguin, in various experiments some of these substances even showed an inhibitory effect. Furthermore, it was found that different target cells show different reactions to the same lysosomatropically active substance.
  • the object of the present invention was to improve the import of nucleic acid into higher eukaryotic cells.
  • import or “transfer”, in addition to the entry of the nucleic acid complexes into the cell through the cell membrane, also means the localization of the complexes or the nucleic acid released therefrom within the cell until one is reached for their expression understood the appropriate location.
  • the higher eukaryotic cells are familiar to the person skilled in the art; Yeast are not counted (Watson et al., 1987).
  • viruses manage their entry into the eukaryotic host via mechanisms which in principle correspond to those of receptor-mediated endocytosis.
  • Virus infection based on this mechanism generally begins with the binding of virus particles to receptors on the cell membrane. The virus is then internalized into the cell. This internalization process follows a common route, corresponding to the entry of physiological ligands or macromolecules into the cell:
  • the receptors on the cell surface initially arrange themselves in groups in order to form a so-called "coated pit", after which the membrane turns inside and forms a vesicle surrounded by an envelope. After this vesicle has got rid of its clathrin shell, it is acidified inside with the help of a proton pump located in the membrane.
  • viruses This causes the virus to be released from the endosome.
  • the virus has a lipid envelope or not, two types of release of the virus from the endosome have been considered:
  • so-called "naked” viruses eg adenovirus, poliovirus, rhinovirus
  • the low pH value causes conformational changes in virus proteins. This exposes hydrophobic domains that are not accessible at a physiological pH. These domains thus acquire the ability to interact with the endosome membrane and thus to release the virus genome from the endosome into the cytoplasm.
  • Viruses with a lipid envelope eg Vesicular Sto atitis Virus, Semliki Forest Virus, Influenza Virus
  • a lipid envelope eg Vesicular Sto atitis Virus, Semliki Forest Virus, Influenza Virus
  • Viruses that enter the cell using this mechanism have certain molecular peculiarities that allow them to break open the endosome membrane in order to gain entry into the cytoplasm.
  • viruses e.g. the enveloped viruses Sendai, HIV and some Moloney leukemia virus strains, or the non-enveloped viruses SV40 and Polyoma do not require a low pH environment for their entry into the cell, can either bring about a fusion with the membrane directly on the cell surface ( Sendai virus, possibly HIV) or they can trigger mechanisms to break open the cell membrane or to pass through it. It is believed that pH-independent viruses can also use the endocytosis route (McClure et al., 1990).
  • the present invention thus relates to a composition for the transfection of higher eukaryotic cells with a complex of nucleic acid and nucleic acid-affine substance, which is optionally coupled to an internalization factor for these cells.
  • the composition is characterized in that it contains an agent which has the ability, per se or as part of the nucleic acid complex, to be taken up into the cells to be transfected and the content of the endosomomes in which the complex after entering the cell is localized to release into the cytoplasm.
  • endosomolytic agent This agent will hereinafter be referred to as "endosomolytic agent”.
  • the ability of the endosomolytic agents to be absorbed into the cells to be transfected and to release the content of the endosomomes in which they are located after entering the cell into the cytoplasm is referred to below as the "uptake function".
  • This uptake function consists of the ability to be internalized into the cell actively, via receptor-dependent endocytosis mechanisms, or passively, via the liquid phase or as part of the nucleic acid complex, and the ability to break up endosomes, which are generally called “ endosomolytic activity "or" endosomolysis "is called.
  • the endosomolytic agent is a virus. In another embodiment, the endosomolytic agent is a virus component.
  • the virus or the virus component which are used in these embodiments of the invention are referred to below as "free virus (component)".
  • the effect of an increasing dose of adenoviruses on the gene transfer capacity of a constant amount of transferrin-polylysine conjugate in HeLa cells was investigated, the luciferase gene being used as the reporter gene.
  • the adenovirus enhancement of gene transfer reached a maximum at 1x104 virus particles per cell, a number which corresponds to the approximate number of adenovirus receptors per HeLa cell.
  • the up to 2000-fold increase in luciferase expression compared to the expression achieved with the transferrin-polylysine conjugates alone corresponded to the higher virus dose.
  • the capacity limiting amounts of conjugate-DNA complexes were examined in the presence of a constant dose of adenovirus.
  • the uptake of adenoviruses into the cells was found to enhance gene transfer mediated by the transferrin-polylysine over a wide range of doses of DNA.
  • the maximum level of gene expression achieved by the conjugate-DNA complexes corresponded to the level achieved with 100 times less DNA when adenoviruses were used to enhance transfection efficiency.
  • adenovirus The effect of adenovirus on gene transfer was examined for both uncomplexed and DNA complexed with polylysine or with transferrin-polylysine conjugates (Fig. 3A). According to this analysis, the presence of adenovirus during transfection only slightly increased the transfer of naked, non-complexed DNA. In sharp contrast to this, the transfer of DNA complexed with polylysine or with transferrin-polylysine conjugates was enhanced by additions of adenovirus, this effect occurring much more strongly with the transferrin-polylysine conjugates.
  • transferrin-polylysine-DNA or polylysine-DNA complexes at low temperature without internalization makes it possible to remove excess complex in the liquid phase before treatment with the adenovirus (FitzGerald et al., 1983). If this was done, the transport of the receptor-bound transferrin-polylysine-DNA complexes was significantly increased by adding adenovirus particles, which was not the case for the polylysine-DNA complexes. It is therefore the entry of the DNA into the cell via receptor-mediated endocytosis that is specifically amplified.
  • adenoviruses had no effect in this cell line, while in the parental cell line into which the gene had been introduced by means of transferrin-polylysine conjugates, they caused a marked increase in gene expression. This finding made it clear that the adenovirus influences events that take place before transcription, that its amplifying effect on the gene import thus acts at the level of the gene import and not at the level of the gene expression (FIG. 5).
  • Cystic fibrosis cell line (CFT1) cells showed moderate luciferase gene expression after treatment with transferrin-polylysine-DNA complexes alone.
  • the level of expression was significantly increased by treatment with the adenovirus dl312.
  • KB cells treated with the transferrin-polylysine-DNA complexes showed luciferase gene expression, which was barely above the background level, despite the presence of transferrin receptors.
  • treatment with adenovirus dl312 caused clearly detectable luciferase activities in these cells.
  • Treatment with adenoviruses had a similar effect on HeLa cells, although this effect was even stronger in these cells.
  • the fact that the degree of amplification varies significantly between different target cells could be due to the fact that this effect is both a function of the number of virus Receptors, for example the adenovirus receptors, of a certain cell type and also the number of transferrin receptors.
  • the nucleic acid-affine substance is preferably an organic polycation, which is preferably conjugated to an internalization factor.
  • DNA which is complexed only with nucleic acid-affine substance, ie without an internalization factor, can be introduced into the cell in the presence of free virus.
  • the complexes consisting of nucleic acid and nucleic acid-affine substance, can be taken up via the liquid phase if the concentration of the complexes is correspondingly high.
  • nucleic acid-affine substance in addition to its ability to produce the extensive electroneutrality of the complex and to compress the nucleic acid into a compact structure, has a sufficient ability to bind to the cell surface in order to penetrate into the cell together with the virus, this can be dispensed with to increase the absorption capacity by covalently binding an internalization factor to the nucleic acid-affine substance in order to deliver the complex into the cell via receptor-mediated endocytosis.
  • Some cells have a relatively high affinity for certain nucleic acids Substances so that the conjugates of nucleic acid and compound factor are absorbed into the cell without the involvement of an internalization factor. This applies, for example, to hepatocytes, which were found in the context of the present invention to take up DNA-polylysine complexes.
  • the endosomolytic agent is a virus that is bound to the nucleic acid-affine substance and that has the ability to enter the cell as part of the conjugate / nucleic acid complex and the content of the endosomes in which the complex after entering the cell is localized to release into the cytoplasm.
  • the endosomolytic agent is a virus component which is bound to a nucleic acid-affine substance and which has the ability to enter the cell as part of the conjuga / nucleic acid complex and the content of the endosomes in which the complex is found localized at the entrance to the cell to release into the cytoplasm.
  • Viruses or virus components bound to a nucleic acid binding domain are referred to below as "viral conjugates" regardless of the type of binding.
  • the viral conjugates which are also the subject of the present invention, contain the virus or the virus component as an integral part of their functional construct and combine the advantages of vector systems based on
  • the viral conjugates according to these embodiments of the invention have the advantage of circumventing the basic limitation inherent in the known bifunctional conjugate system for gene transfer via receptor-mediated endocytosis by having a specific mechanism for their release from the cell vesicle System enables.
  • the viral conjugates according to the invention represent a fundamental conceptual departure from the recombinant viral vectors in that the foreign DNA to be transported is carried on the outside of the virion. As a result, very large gene constructs can also be transported into the cell with the aid of the conjugates according to the invention, with no restrictions with regard to the sequence.
  • a virus to be used as a free or bound virus (part) as a virus component in the context of the present invention is defined by its uptake function.
  • suitable viruses are those which have the ability to penetrate the cell via the receptor-mediated endocytosis during the transfection of the cells with the nucleic acid complex and to induce their release - and thus the release of the nucleic acid - from the endosome into the cytoplasm. Without wishing to be bound by this theory, this mechanism could benefit the nucleic acid complexes imported into the cell insofar as they, if they get into the same endosomes during internalization as the viruses, together with the viruses from the endosomes into the cytoplasm to get promoted.
  • nucleic acid complexes contain the virus in bound form, they benefit from the endosomolytic activity of the virus and are transported from the endosomes into the cytoplasm.
  • the fusion between endosomes and lysosomes and thus the enzymatic degradation normally occurring in these cell organelles should be avoided.
  • viruses and higher eukaryotic cells that they can penetrate are e.g. Fields and Knipe, 1990, removable.
  • the susceptibility of a given cell line to transformation by a virus used in the form of a free virus to facilitate entry of nucleic acid complexes into cells depends on the presence and number of surface receptors for the virus on the target cell.
  • the cell surface receptor for adenovirus methods for determining its number on HeLa and KB cells have been described by Svensson, 1990 and Defer, 1990.
  • Viruses which are suitable for the composition according to the invention and whose absorption function takes place at the beginning of the infection via receptor-mediated endocytosis include, on the one hand, viruses without a lipid envelope, such as adenovirus, poliovirus, rhinovirus, and, on the other hand, the envelope viruses vesicular stomatitis virus, Semliki Forest Virus, Influenza virus; pH-dependent strains of Moloney virus are also suitable.
  • viruses for use in the present invention include adenovirus, sub-group C, type 5, Semliki Forest Virus, Vesicular Stomatitis Virus, Poliovirus, Rhinoviruses and Moloney Leukemia virus strains.
  • transcriptase has the advantage that transfection in the presence of such a virus does not result in the formation of viral DNA in the cell.
  • rhinovirus HRV2 a member of the picornavirus group, increases the expression of a reporter gene. The performance of the rhinovirus has been demonstrated both in free form and in the form of virus conjugates.
  • viruses - provided that they are taken up into the cell and release the content of the endosomes into which they come - are understood to mean not only the wild types but also mutants which, due to one or more mutations, have functions of the wild type which are of the recording function are different, especially the replication ability, have been lost.
  • Such mutants are produced by mutations or deletions in virus-protein regions which are responsible for replication functions and are unnecessary for the uptake function and which can be complemented by the packaging line, using conventional mutagenesis methods. These include, for example in the case of adenovirus, ts mutants (temperature-sensitive mutants), E1A and EIB mutants, mutants which have mutations in MLP-driven genes (Berkner, 1988) and mutants which have mutations in regions of certain capsid proteins. Virus strains which have corresponding natural mutations are also suitable.
  • viruses can be examined, for example, with the aid of plague assays known from the literature, in which cell cultures are coated with suspensions of different virus concentrations and the number of lysed cells, which can be seen from Plagues, is found (Dulbecco, 1980).
  • So-called defective viruses are also suitable for use in the context of the present invention, these are viruses which lack the function required for autonomous virus replication in one or more genes and for which they need helper viruses.
  • Representatives of this group are DI ("defective interfering particles"), which are derived from the infectious standard virus, have the same structural proteins as the standard virus, have mutations and require the standard virus as a helper virus for their replication (Huang, 1987; Holland, 1990 ).
  • the satellite viruses are also a representative of this group (Holland, 1990).
  • Another group is the class of parvoviruses called "adeno-associated virus" (Berns, 1990). Since the uptake cycles of many viruses into the cell have not yet been fully elucidated, it is believed that there are other viruses that have the endosomolytic activity necessary for their suitability for use in the present invention.
  • Attenuated live vaccines (Ginsberg, 1980) or vaccine strains are also suitable in the context of the present invention.
  • viruses in the context of the present invention includes inactivated viruses, e.g. by chemical treatment, such as formaldehyde treatment, by UV radiation, by chemical treatment, combined with UV radiation, e.g. Psoralen / UV or bromodeoxyuridine / UV treatment, viruses inactivated by gamma radiation or neutron bombardment.
  • chemical treatment such as formaldehyde treatment
  • UV radiation e.g. Psoralen / UV or bromodeoxyuridine / UV treatment
  • viruses inactivated by gamma radiation or neutron bombardment e.g. by UV radiation, by chemical treatment, combined with UV radiation, e.g. Psoralen / UV or bromodeoxyuridine / UV treatment, viruses inactivated by gamma radiation or neutron bombardment.
  • TZBLATT Inactivated viruses such as those used for vaccines, can be produced using standard methods known from the literature (Davis and Dulbecco, 1980, Hearst and Thiry, 1977) and tested for use to increase the import of DNA complexes.
  • adenovirus preparations were inactivated using a conventional UV sterilization lamp or with formaldehyde and it was surprisingly found that the extent of inactivation of the viruses was substantially greater than the decrease in the gene transfer effect which was achieved. if adenovirus has been added to the transfection medium.
  • virus components is understood to mean parts of viruses, for example the protein portion freed from nucleic acid (the empty virus capsid which can be prepared using recombinant methods, see for example Ansardi et al., 1991, Urakawa et al., 1989), proteins which are obtained in the fractionation or peptides which possess the endosomolytic ability of the intact virus which is essential for the uptake function.
  • virus components can also be produced synthetically, depending on their size, either by peptide synthesis or using recombinant methods. In the context of the present invention it could be shown that adenovirus proteins which are conjugated to polylysine via biotin / streptavidin can increase the gene transfer.
  • influenza virus hemagglutinin The N-terminal sequence of the influenza virus hemagglutinin HA2 subunit is responsible for the release of the virus from the endosome. It has been shown that peptides consisting of 20 amino acids of this sequence can fuse and partially break up / destroy lipid membranes (Wharton et al., 1988). In the present invention, authentic and modified influenza apeptides have been successfully used in various embodiments.
  • envelope proteins from retroviruses for example HIV gp41 (Rafalski et al., 1990) or parts of these virus proteins.
  • viruses that have the inherent ability to enter cells is only one aspect of the present invention.
  • Viruses or virus components that do not have the inherent ability to bind to and penetrate the cell are preferably used in the form of the viral conjugates defined above. Coupling to a DNA binding domain, e.g. a polycation ensures that the virus or the virus component has a strong affinity for DNA molecules, so that it is complexed and transported into the cell as part of the DNA complex, which also contains an internalizing factor / DNA binding doane conjugate. In addition to the transport effect achieved with it
  • a simple screening assay can be used to determine whether a given virus or a virus component is suitable for an uptake function within the meaning of the present invention and thus for use in enhancing gene transfer.
  • the target cells are brought into contact with a DNA complex in the presence or absence of the virus.
  • the amount of DNA complex that is released into the cytoplasm can then be determined simply by detecting a marker gene product, for example luciferase. If the presence of the virus results in the DNA complex being absorbed into the cell to a greater extent than without the virus and released into the cytoplasm, this can be attributed to the uptake function of the virus.
  • test virus in terms of its uptake function with a virus which is known to have a suitable uptake function, for example with adenovirus, subgroup C, type 5.
  • Tests of this type can also be applied to viral conjugates, where additional parameters, such as different internalizing actuator conjugates, can be subjected to such tests in different amounts.
  • those of ordinary skill in the art can perform assays of this type, optionally in conjunction with other tests such as liposome permeability assays (“Liposome Leakage Assays”), easy to apply to virus components or other agents with potential endosomolytic activity to examine them for their ability to increase gene expression.
  • liposome permeability assays liposome permeability assays
  • virus If intact viruses are used, it is expedient to check whether the virus is capable of replication, in parallel to the preliminary tests in which the virus is examined for its ability to enhance gene transfer. In the case of cytopathic viruses or in the case of viruses which noticeably impair the growth of the host cells, the examination for replication ability is carried out with the aid of plague assays (cf. above). For other viruses, detection methods are used specifically for the respective virus, e.g. the hemagglutionation test or chemical-physical methods (electron microscopic).
  • viruses which can be produced in high titer, which are stable, which have low pathogenicity as a wild type and in which a targeted deactivation of the replication functions is possible, in particular adenoviruses. If a particular cell population is to be transfected, viruses are preferred which specifically infect this cell population. In the event that different cell types are to be detected from the transfection, viruses can be used which are infectious for a wide range of cell types.
  • viruses or virus components are considered for the therapeutic application of the present invention in vivo, in which safety risks, in particular with regard to the replication of the virus in the cell and the recombination of virus DNA with host DNA, are largely minimized are.
  • the entry mechanism of viruses infecting animals other than humans can be used to enhance the uptake and release of DNA in higher eukaryotic cells, particularly human cells, provided the virus has the ability to disrupt endosomes in the cells .
  • Members of the adenovirus family have been isolated from bird species, amphibians, and various other animals (see, e.g., Laver et al., 1971; Bragg et al., 1991; Akopian et al., 1991; Takase et al., 1990; Khang and Nagaraji, 1989 , and Reece et al., 1987).
  • Amphibian, bird, cattle, dog, mouse, sheep, pig and monkey adenoviruses, as well as human adenoviruses, are available from the American Type Culture Collection, Rockville, Maryland (see American Type Culture Collection Catalog of Animal Viruses and Antisera, Chlamydae and Rickettsiae, 6th ed., 1990, C. Bück and G. Paulino eds., pp. 1-17).
  • a virus e.g. an adenovirus
  • a distant species Potential advantages of using a virus, e.g. an adenovirus, of a distant species would be reduced toxicity in the target cells (e.g., chicken or frog adenovirus would not be expected to replicate in mammalian cells or initiate expression of early genes), in comparison to human adenovirus reduced risk for the researcher of this virus from a distant species and a reduced interference by antibodies against the human or mouse adenovirus.
  • the absence of interference from human or mouse antibodies is particularly important when the viruses are used for gene therapy in humans or in the mouse.
  • the chicken adenovirus CELO (Chick Embryo Lethal Orphan Virus) shows no reactivity with antibodies that recognize the main group epitopes of the adenoviruses that infect mammalian cells.
  • CELO viruses can be grown in eggs with embryo development to obtain large amounts of virus (0.5 mg / egg; Laver et al., 1971).
  • CELO-polylysine conjugates enhance the DNA transport in HeLa cells to an extent comparable to the human adenovirus dl312. The use of CELO conjugates to enhance DNA transport is therefore very promising for human gene therapy.
  • Viruses from distant species are preferably used as components of viral conjugates in combination complexes (as defined in the context of the present invention).
  • the binding of the virus to the nucleic acid binding domain can be covalent or non-covalent, the latter e.g. via a biotin-streptavidin bridge or, in the event that the virus has regions on its surface proteins which are acidic and can therefore bind to a polycation, via an ionic bond.
  • virus components in the conjugates according to the invention with endosomolytic activity are the empty virus capsids or viral peptides.
  • the binding of the virus component to the nucleic acid binding domain can be covalent, e.g. by chemical coupling of the viral peptide with polylysine, or non-covalently, e.g. ionic in the event that the virus component has acidic residues to bind to a polycation.
  • the ratio of virus or virus component to the nucleic acid-affine substance can be varied.
  • influenza-hemagglutinin-peptide-polylysine conjugate it was found within the scope of the present invention that the gene transfer can be increased to a greater extent if the content of viral peptide in the conjugates was higher.
  • the present invention relates to methods for producing the conjugates according to the invention from the virus (component) and nucleic acid-affine substance.
  • the viral conjugates (like the internalization factor or polycation conjugates) can be prepared by coupling the components or, if the virus component and the polycation are polypeptides, by a recombinant route with respect to Manufacturing methods are referred to the disclosure of EP 388 758.
  • the coupling of virus, virus proteins or peptides with polyamine compounds by chemical means can be carried out in a manner known per se for the coupling of peptides, whereby, if necessary, the individual components are provided with linker substances before the coupling reaction (this measure is necessary if by no functional group suitable for the coupling, for example a mercapto or alcohol group, is available in advance
  • the linker substances are bifunctional compounds which are first reacted with functional groups of the individual components, after which the modified individual components are coupled.
  • the coupling can be done via
  • Disulfide bridges that can be split again under reducing conditions (e.g. when using succinimidylpyridyldithiopropionate (Jung et al., 1981).
  • SPDP Succinimidylpyridyldithiopropionat
  • adenovirus was coupled to polylysine using various methods.
  • virus or the virus component has suitable carbohydrate chains, they can with the nucleic acid-affine substance over one or more Carbohydrate chains of the glycoprotein are linked together.
  • Another preferred method for producing the conjugates according to the invention is the enzymatic coupling of the virus or the virus component to a substance affine to nucleic acid, in particular a polyamine, by means of a transglutaminase.
  • transglutaminases comprises several different enzymes, which are found in the epidermis (epidermal transglutaminase), in the blood (factor XIII) and in the cells of different tissues (tissue transglutaminase) (Folk, 1985).
  • Transglutaminases catalyze the formation of ⁇ - ( ⁇ -glutamyl) lysine bonds in the presence of Ca ++ and with the elimination of NH3.
  • a prerequisite for this is that corresponding glutamines and lysines are present on proteins which can be converted by the enzyme.
  • (poly) amines such as ethanolamine, putreszine, spermine or spermine can also serve as the substrate (Clarke et al., 1959).
  • glutamine or lysine of a protein or a polyamine can be converted by the enzyme.
  • numerous cell proteins such as cytokeratins (Zatloukal et al., 1989), tubulin, cell membrane proteins and also surface proteins of influenza viruses (Iwanij, 1977) can bind polyamines by means of transglutaminase.
  • Adenoviruses can be coupled. It was found that the coupling in the presence of
  • Glycerin can be done. This offers the
  • a virus preparation e.g. a
  • Coupling can be used.
  • Plasmid DNA could be complexed one by one
  • Another preferred method in the context of the present invention for producing the conjugates according to the invention is to couple the virus or the virus component to the polycation via a biotin-protein bridge, preferably a biotin-streptavidin bridge.
  • binding to biotin can also take place via avidin.
  • virus (component) and polylysine it is possible to establish the link between virus (component) and polylysine, on the one hand by biotinylating the virus and, on the other hand, conjugating an anti-biotin antibody with polylysine and establishing the link between virus and polylysine via the biotin / antibody linkage, commercially available polyclonal or monoclonal antibodies against biotin can be used.
  • the link between the virus and polylysine can also be established by coupling polylysine with a lectin that has affinity for a surface virus glycoprotein, the binding in such a conjugate being via the link between the lectin and the glycoprotein. If the virus does not have suitable carbohydrate side chains, it can be modified accordingly.
  • a virus can also be bound to a substance with an affinity for nucleic acids by first modifying its surface with a non-virus antigen (eg digoxigenin DIG, available from Boehringer Mannheim; or with biotin) and the connection between the modified virus and the nucleic acid.
  • a non-virus antigen eg digoxigenin DIG, available from Boehringer Mannheim; or with biotin
  • affine substance is produced via an antibody that binds to this antigen.
  • T Biotin is the most unspecific and therefore the most widely applicable method, the biotin-mediated binding represents a very strong non-covalent binding.
  • the enzymatic reaction with transglutaminase has the advantage that it can also be carried out on the smallest scale.
  • Chemical coupling is generally used when larger amounts of conjugate are to be synthesized; this method is generally also most expedient when virus proteins or peptides are to be coupled. If inactivated viruses are used, the inactivation is generally carried out before the coupling, provided that the coupling is not impaired by the inactivation.
  • a virus e.g. Adenovirus or an endosomolytic component thereof, having accessible binding domains, e.g. acidic domains for binding to a polycation
  • the binding of the virus or the virus component to the polycation can also be ionic.
  • the positive charges of the polycation which may be conjugated with an internalization factor, are partially neutralized by the acidic domain of the virus (component), the rest of the positive charges are essentially neutralized by the nucleic acid.
  • an intercalating substance is used as the nucleic acid-affine substance, it is modified with a linker suitable for the respective coupling of virus (component), e.g. for coupling with transglutaminase with spermine, or with a bifunctional group which is competent for chemical coupling, e.g. an active ester.
  • a linker suitable for the respective coupling of virus e.g. for coupling with transglutaminase with spermine
  • a bifunctional group which is competent for chemical coupling, e.g. an active ester.
  • the ratio of virus (component): nucleic acid Affine substance can vary, it is usually determined empirically, for example by conjugating a constant amount of virus (component) with different amounts of polylysine and selecting the optimal conjugate for transfection.
  • the virus component e.g. an endosomolytic viral peptide
  • modified to bind directly to DNA the peptide itself can have a DNA binding domain which can be obtained by producing the peptide by means of peptide synthesis, a section of positively charged amino acids being provided, preferably by extending the peptide, particularly preferably at the C-terminus.
  • the endosomolytic agent is a non-viral, optionally synthetic peptide.
  • a peptide of this type is preferably included in the composition according to the invention in such a way that it is ionic to the nucleic acid-affine substance, e.g. is bound to polylysine in the case of DNA / internalizing factor-polylysine complexes.
  • the incorporation of the endosomolytic peptide into the nucleic acid complexes is brought about by the peptide being bound via its acidic amino acid residues to the positively charged nucleic acid binding domain, preferably polylysine.
  • the binding to polylysine can also take place by the methods described here for the binding of peptides to polylysine.
  • this peptide can be used with a suitable terminal amino acid as
  • ATZBLATT "Stalk” can be modified for conjugation.
  • nucleic acid complexes Another way of incorporating non-viral endosomolytic peptides into the nucleic acid complexes is to provide them with sequences that bind to DNA. The location of such a sequence must be such that it does not interfere with the endosomolytic activity of the peptide. Therefore e.g. Peptides whose N-terminus is responsible for this activity are extended at the C-terminus with DNA-binding sequences. Extensions of this type can be homologous or heterologous cationic oligopeptides, e.g. an oligolysin tail, or a natural DNA binding domain, e.g. a peptide derived from a histone.
  • DNA binding sequences which form the endosomolytic peptide and are approximately 10-40 amino acids.
  • This embodiment of the invention offers the possibility of a higher ratio of endosomolytic sequence: DNA binding sequence than in peptide conjugates which contain larger proportions of polycations in order to achieve a higher performance of the complexes.
  • non-viral endosomolytic peptides should meet the following requirements:
  • the permeability of lipid membranes caused by the peptide should preferably be higher at low pH (5-6) than at pH 7. Furthermore, the broken membrane areas should be large enough to allow the passage of large DNA Allowing complexes (small pores are not sufficient).
  • in vitro tests can be carried out in which the peptides are in free or bound form and / or incorporated into one DNA complex can be applied. Such tests can be liposome or erythrocyte permeability tests
  • Peptides that rupture membranes generally contain amphipathic sequences, namely a hydrophobic side that can interact with the lipid membrane and a hydrophilic side that stabilizes the aqueous phase at the point where the membrane ruptures.
  • membrane breaking peptides in nature, usually small peptides or peptide domains of large polypeptides.
  • Such peptides can be classified according to their function in the natural context, namely either peptides breaking up in membranes (e.g. peptides from naked viruses) and / or membrane-fusing peptides (e.g. peptides from enveloped viruses). Both classes of peptide sequences can be useful for disrupting endosomes associated with synthetic peptides. Most natural peptides can form amphipathic ⁇ helices.
  • An amphipathic peptide sequence is selected from the group of naturally occurring or artificial peptides. Peptides of this type belong to the prior art; an overview of examples is given in Table 2. If necessary, acidic residues (Glu, Asp) are introduced in order to make the activity of the peptide, membrane rupture, more pH-specific (e.g. the double acid mutant of the influenza hemagglutinin peptide of the designation P50 according to Example 37). If necessary, acidic residues can also be introduced to facilitate the binding of the peptide to polylysine. One way to provide such a polycation binding domain can be to add C-terminal acidic extensions, e.g. to provide an oligo-glu tail.
  • Endosomolytic peptides can also be obtained by using naturally occurring and artificial sequences
  • the length of the peptide sequence can be critical with respect to the amphipathic helix; increasing the stability of short domains derived from natural proteins and lacking the context of the stabilizing protein can be achieved by extending the helix.
  • homodimers, heterodimers or oligomers can be formed; in the examples of the present invention it was shown that a P50 dimer has a much higher activity than the monomer.
  • the inventors have shown the effect of synthetic peptides on DNA uptake using transferrin-polylysine conjugates.
  • Several different peptides were synthesized, their ability to render liposomes and erythrocytes permeable, and their effects on luciferase expression in TIB 73 cells and in NIH 3T3 cells tested.
  • a non-peptide amphipathic substance In a further embodiment of the invention that is endosomolytic agent a non-peptide amphipathic substance.
  • the requirements that such a substance has to meet in order to be suitable for use in the context of the present invention are essentially the same as for the amphipathic peptides, namely the ability to be integrated into the nucleic acid complex, pH specificity, Etc.
  • the invention relates to complexes which are taken up in higher eukaryotic cells, containing nucleic acid and a conjugate which has the ability to form a complex with nucleic acid, for introducing nucleic acid into higher eukaryotic cells.
  • the complexes are characterized in that they contain a conjugate which consists of a nucleic acid-affine substance and an endosomolytic agent which is bound to the nucleic acid-affine substance and has the ability to enter the cell as part of a conjugate / nucleic acid complex to be absorbed and to release the contents of the endosomes, in which the complex is located after entering the cell, into the cytoplasm.
  • nucleic acid complexes which are used in the context of the present invention are preferably those in which the nucleic acid is complexed with a substance affine to nucleic acid in such a way that the complexes are essentially electroneutral.
  • the endosomolytic agent is a virus or a virus component which is or are covalently bound to a polycation.
  • the endosomolytic conjugates - in addition to conjugates in which endosomolytic agents are ionically bound to a DNA binding domain - by definition also include endosomolytic agents which bind directly to DNA, for example via their basic extension, although "conjugates" of this type strictly speaking, not obtained by conjugation, ie by connecting two components together.
  • endosomolytic agents of this type as constituents of the composition according to the invention is independent of whether they were synthesized by conjugation of an endosomolytic agent and a DNA binding domain or whether a DNA binding domain was originally present in the endosomolytic agent.
  • the complexes contain, in addition to the endosomolytic conjugate, a further conjugate in which a nucleic acid-affine substance, in the case of an endosomolytic polycation conjugate, is generally the same as that of the conjugate, is coupled to an internalization factor with affinity for the target cell .
  • a nucleic acid-affine substance in the case of an endosomolytic polycation conjugate, is generally the same as that of the conjugate, is coupled to an internalization factor with affinity for the target cell .
  • Another application of this embodiment is that in which a virus component, for example a naturally occurring, optionally modified peptide, a non-viral, optionally synthetic endosomolytic peptide or a virus of a distant species are used which do not have the ability to inherently in the too penetrate transfecting cells.
  • a virus component for example a naturally occurring, optionally modified peptide, a non-viral, optionally synthetic endosomolytic peptide or a virus of a distant species are used which do not have the ability to inherently in the too penetrate transfecting cells.
  • a virus component for example a naturally occurring, optionally modified peptide, a non-viral, optionally synthetic endosomolytic peptide or a virus of a distant species
  • the combination complexes of cells are either by binding to the surface receptor specific for the internalizing factor or, in the case of using a virus or a virus component, by binding to the virus receptor or by binding to both receptors the pathway of receptor-mediated endocytosis was added.
  • the endosomolytic agents are released from the endosomes, the DNA contained in the complexes is also released into the cytoplasm and thereby escapes lysosomal degradation.
  • viruses, virus components or non-viral endosomolytic agents as components of endosomolytic conjugates in the DNA complexes has the following advantages:
  • internalization factor is understood to mean ligands or fragments thereof which are internalized after binding to the cell via endocytosis, preferably receptor-mediated endocytosis, or factors whose binding / internalization takes place via fusion with cell membrane elements.
  • Suitable internalization factors include the ligands transferrin (Klausner et al., 1983), conalbumin (Sennett et al., 1981), asialoglycoproteins (such as asialotransferrin, asialorosomucoid or asialofetuin), (Ashwell et al., 1982), lectins (Goldstein et al ., 1980 and Shardon, 1987), or substances which contain galactose and are internalized via the asialoglycoprotein receptor; mannosylated glycoproteins (Stahl et al., 1987), lysosomal enzymes (Sly et al., 1982), LDL (Goldstein et al., 1982), modified LDL (Goldstein et al., 1979), lipoproteins that are receptors in the Cell are taken up (apo B100 / LDL); viral proteins such as the HIV protein gpl20; Antibodies (Mellman et al.,
  • immunoglobulins or fragments thereof as ligands for the Fc receptor or anti-immunoglobulin antibodies which respond to SIgs ("Surface Immunoglobulins").
  • the ligands can be of natural or synthetic origin (see Trends Pharmacol. Sei-, 1989, and the references cited therein).
  • the broad applicability of the invention with regard to the internalization factor was demonstrated or an additional internalization factor in the combination complexes based on human and mouse transferrin-polylysine conjugates, asialofetuin-polylysine conjugates, galactose-polylysine conjugates, wheat germ agglutinin-pL conjugates, the T cell-specific gpl20-pL and antiCD7-pL conjugates, of LDL-pL conjugates, Ig-pL and anti-Ig-pL conjugates as well as DNA-polylysine complexes that do not contain an internalization factor.
  • the performance of the viral conjugates according to the invention was shown on the basis of complexes of DNA and polylysine-conjugated virus (or virus component) which did not contain any additional internalizing factor-connecting factor conjugate.
  • preliminary tests can determine whether, in the case of using free virus as an endosomolytic agent, the use of an internalization factor or whether the endosomolytic agent uses a virus or a virus component or a non-viral peptide as part of an endosomolytic conjugate is, an "additional" connection factor enables or improves the uptake of nucleic acid complexes.
  • Such tests include parallel transfections with nucleic acid complexes once without (additional) internalization factor, e.g. in the case of viral co ugates with complexes consisting of nucleic acid and viral conjugate, and once with complexes in which the nucleic acid with another conjugate containing another internalization factor for which the target cells have a receptor.
  • an internalization factor or an additional internalization factor is used, ie if a combination complex is used, this becomes Defined above all by the target cells, for example by certain surface antigens or receptors that are specific for a cell type and thus enable a directed import of the nucleic acid into this cell type.
  • Suitable nucleic acid-affine substances within the scope of the present invention are e.g. homologous organic polycations such as polylysine, polyarginine, polyornithine or heterologous polycations with two or more different positively charged amino acids, where these polycations can have different chain lengths, furthermore non-peptide synthetic polycations such as polyethyleneimine.
  • Suitable nucleic acid-affine substances are also natural DNA-binding proteins polycation, see characters such as histones or protamines or analogs or fragments thereof, as well as spermine or spermidines.
  • the length of the polycation is not critical insofar as the complexes are essentially electroneutral with regard to the preferred embodiment.
  • the preferred range of polylysine chain length is from about 20 to about 1,000 lysine monomers. However, there is no critical length for the polycation for a given DNA length. If the DNA consists of 6,000 bp and 12,000 negative charges, the amount of polycation per mole of DNA can e.g. his:
  • nucleic acid-affine substances as Part of the F. conjugates are intercalating substances such as ethidium dimers, acridine or intercalating peptides containing tryptophan and / or tyrosine and / or phenylalanine.
  • the nucleic acid to be imported into the cell is generally determined first.
  • the nucleic acid is primarily defined by the biological effect to be achieved in the cell, in the case of use in the context of gene therapy by the gene to be expressed or the gene segment, e.g. for the purpose of substituting a defective gene or by targeting a gene to be inhibited.
  • the nucleic acids to be transported into the cell can be DNAs or RNAs, with no restrictions with regard to the nucleotide sequence.
  • the cell to be inserted into the DNA preferably codes for an immunomodulating substance, for example a cytokine such as IL-2, IL-4, IFN-gamma, TNF- ⁇ .
  • an immunomodulating substance for example a cytokine such as IL-2, IL-4, IFN-gamma, TNF- ⁇ .
  • cytokines for example IL-2 and IFN-gamma
  • Another gene that is useful for insertion into tumor cells is the "multi drug resistance gene" (mdr).
  • midr multi drug resistance gene
  • transferrin-polylysine and low-density lipoprotein conjugates were successfully used together with adenovirus conjugates for the transfection of tumor cells (melanoma cells).
  • preliminary tests can determine which ligand is suitable for the specific application for the respective tumor cell type. It is also possible to introduce two or more different nucleic acid sequences into the cell, for example a plasmid containing cDNAs, coding for two different proteins, under the control of suitable regulatory sequences, or two different plasmid constructs containing different cDNAs.
  • Therapeutically active inhibiting nucleic acids which are introduced into the cell for the purpose of inhibiting specific gene sequences include gene constructs from which antisense RNA or ribozymes are transcribed. It is also possible to use oligonucleotides, e.g. Antisense oligonucleotides to be introduced into the cell. Antisense oligonucleotides preferably comprise 15 nucleotides or more. The oligonucleotides can optionally be multimerized. Ribozymes are preferably used as part of a gene construct that contains stabilizing gene elements, e.g. contains tRNA gene elements, introduced into the cell. Gene constructs of this type are disclosed in EP A 0387 775.
  • Inhibiting nucleic acids and their mechanisms of action are familiar to the person skilled in the art; in this regard, reference is made to the review articles by Helene and Toulme, 1990 and Takayama and Inouye, 1990, as well as the references cited therein.
  • genes with other inhibitory effects can also be used.
  • examples include genes that code for viral proteins that have so-called transdominant mutations (Herskowitz, 1987). Expression of the genes in the cell yields proteins that dominate the corresponding wild-type protein and in this way the cell that acquires cellular immunity protect by preventing virus replication.
  • Transdominant mutations of viral proteins which are required for replication and expression are suitable, for example Gag, Tat and Rev mutants which have been shown to inhibit HIV replication (Trono et al., 1989; Green et al. , 1989; Mali et al., 1989).
  • RNA molecules that contain the binding site for an essential viral protein, e.g. so-called "TAR Decoys” (Sullenger et al., 1990).
  • genes which can be used in the context of somatic gene therapy and which can be introduced into the cell as part of gene constructs with the aid of the present invention are factor VIII (haemophilia A) (see, for example, Wood et al. ' , 1984), factor IX (Hemophilia B), (see e.g. Kurachi et al., 1982), adenosine deaminase (SCID), (see e.g. Valerio et al., 1984), ⁇ -1 antitrypsin (pulmonary emphysema), (see e.g. Ciliberto et al., 1985) or the "Cystic fibrosis transmembrane conductance regulator gene" (see, for example, Riordan et al., 1989). These examples are not limitative.
  • nucleic acid molecules with a size of approximately 0.15 kb (in the case of a tRNA gene containing a ribozyme gene) to approximately 50 kb and above can be transported into the cell; smaller nucleic acid molecules can be used as oligonucleotides. It is obvious that it is precisely because the fact that the present invention is not subject to any restrictions in terms of the gene sequence and that very large gene constructs can also be transported with the aid of the invention.
  • the nucleic acid-affine substance preferably an organic polycationic substance, is determined which ensures the complexation of the nucleic acid; the complexes obtained are preferably essentially electroneutral. If, in addition to the endosomolytic conjugate, the complexes contain a conjugate of an internalization factor and a nucleic acid-affine substance, the cation content of both conjugates is taken into account with regard to the aspect of electroneutrality.
  • nucleic acid was chosen so that the internalizing factor polycation / nucleic acid complexes were largely electroneutral. It has been found that when a portion of the transferrin-polycation conjugates are replaced with non-covalently bound polycations, the amount of nucleic acid taken up into the cell is not reduced; in certain cases, a significant increase in DNA uptake can even be achieved (Wagner et al., 1991a). It was observed that the DNA within the complexes is present in a form condensed into toroidal structures with a diameter of 80-100 nm.
  • the amount of polycation is thus in view of the two The parameters selected as electroneutrality and the achievement of a compact structure, the amount of polycation which results from the charge of the nucleic acid with a view to achieving electroneutrality generally also ensures a compacting of the DNA.
  • the complexes may therefore additionally contain nucleic acid-binding substances in non-covalently bound form, which may be the same or different from the compound factor, that is to say the nucleic acid-affine substance in the conjugates.
  • the complexes include nucleic acid and
  • connection factor conjugate In the event that an endosomolytic, for example a viral, conjugate is used, the nucleic acid is complexed with this conjugate, possibly together with a conjugate of an additional internalization factor.
  • the choice of non-covalently bound "free" nucleic acid-affine substances by type and amount is also matched to the conjugate or conjugates, whereby the connection factor contained in the conjugate must be taken into account above all: is the connection factor, for example, a substance that does not or has only a slight ability to condense DNA, it is generally expedient in view of an efficient internalization of the complexes to use substances which are affinity to DNA and which possess this property to a greater extent.
  • connection factor itself is a nucleic acid-condensing substance and if it already effects a compacting of the nucleic acid that is sufficient with regard to efficient internalization, it is expedient to have a nucleic acid affinity Substance used that causes an increase in expression due to other mechanisms.
  • nucleic acid-affine substances which are suitable in the context of the present invention include compounds with the ability to condense nucleic acid and / or to protect them from undesired degradation in the cell, in particular the substances of polycationic character already mentioned.
  • suitable substances are those which, by binding to the nucleic acid, improve their transcription / expression by improving the accessibility of the nucleic acid to the expression machinery of the cell.
  • An example of such a substance is the chromosomal non-histone protein HMG1, which has been found to have the ability to compact DNA and to express it in the cell.
  • Internalization factor / connection factor / nucleic acid depends primarily on the size of the polycation molecules as well as on the number and distribution of the positively charged groups, criteria based on Size and structure of the nucleic acid (s) to be transported are coordinated.
  • the molar ratio is preferably
  • the amount of the internalizing factor-conjugate portion can be determined with the aid of titrations, which portion can be replaced by free nucleic acid-affine substance. If polycations are used both as a connecting factor and as a free nucleic acid-affine substance, the polycations can be the same or different.
  • a suitable method for determining the ratio of the components contained in the complexes is to first define the gene construct to be imported into the cell and, as described above, a virus or a Find virus component that is suitable for the specific transfection. Then the virus or the virus component is bound to the polycation and complexed with the gene construct. Based on a defined amount of viral conjugate, titrations can be carried out by treating the target cells with this (constant) amount of conjugate and decreasing DNA concentrations, or vice versa. In this way, the optimal ratio of DNA: virus conjugate is determined. When using an additional internalization factor, for example, the procedure is such that the optimal ratio is based on a constant amount of DNA by titrations between virus conjugate and internalizing factor conjugate is determined.
  • the complexes can be prepared by the components i) nucleic acid, ii) viral conjugate, optionally iii)
  • connection factor conjugate and optionally iv) non-covalently bound nucleic acid-affine substance which are in the form of dilute solutions, are mixed. If polycations are used as the connecting factor and at the same time as "free" polycations, it is generally expedient to first produce a mixture of conjugates with free polycations and to combine this mixture with DNA.
  • the optimal ratio of DNA to conjugate (s) and polycations is determined by titration experiments, i.e. in a series of transfection experiments with constant amount of DNA and increasing amount of conjuga (s) / polycation mixture.
  • the optimal ratio of conjugate (s): polycations in the mixture can be determined with the help of routine experiments or by comparing the optimal ratios of those in the
  • the DNA complexes can be produced at physiological salt concentrations. Another possibility is the use of high salt concentrations (about 2 M NaCl) and subsequent adjustment to physiological conditions by slow dilution or dialysis.
  • nucleic acid, conjugate (s), any non-covalently bound free nucleic acid-affine substance is determined in detail in preliminary experiments. In some cases it may prove expedient to first complex the nucleic acid with the conjugate and only then to add the free nucleic acid-affine substance, for example the polycation, for example in the case of transferrin-ethidium dimer conjugates and polylysine.
  • the (additional) internalization factor is transrin and the connection factor is a polycation.
  • Trans errin is to be understood both as the natural transferrin and as those transferrin modifications that are bound by the receptor and transported into the cell.
  • the nucleic acid is taken up in the form of complexes in which internalizing factor-polycation conjugates are complexed with nucleic acid.
  • this is preferably a polycation which is the same or different from the polycation contained in the conjugate.
  • the nucleic acid is internalized in the form of complexes in which, on the one hand, the internalization factor conjugates and, on the other hand, the endosomolytic ones are complexed with nucleic acid.
  • the internalizing factor-polycation conjugates which are used together with free virus or with the viral conjugates in the combination complexes can be prepared chemically or, if the polycation is a polypeptide, recombinantly, with regard to the production methods reference is made to the disclosure of EP 388 758.
  • the conjugates can also be made by adding a glycoprotein, e.g. Transferrin, and the connection factor are connected to one another via one or more carbohydrate chains of the glycoprotein.
  • a glycoprotein e.g. Transferrin
  • the connection factor are connected to one another via one or more carbohydrate chains of the glycoprotein.
  • conjugates produced by means of conventional coupling methods such conjugates are free from modifications which originate from the linker substances used.
  • These conjugates have in the case of glycoproteins which have only one or a few carbohydrate groups suitable for coupling, e.g. Transferrin, also have the advantage that they are precisely defined in terms of their binding site glycoprotein / connection factor.
  • the amount of endosomolytic agent used or its concentration depends on the individual transfection. It is expedient to use the minimum amount of virus or virus component that is necessary to ensure the internalization of the virus and nucleic acid complex and the release from the endosomes.
  • the amount of virus (conjugate) is matched to the respective cell type, taking into account the infectivity of the virus for this cell type.
  • a further criterion is the respective internalization factor-connection factor conjugate, in particular with regard to the internalization factor, for which the target cell has a defined number of receptors.
  • the amount of virus (conjugate) depends on the amount of DNA to be imported.
  • a small amount of virus is generally sufficient for stable transfection, for which only a small amount of DNA is required, while for a transient transfection, for which larger amounts of DNA are required, the amount of virus used is greater.
  • the optimal virus concentration is determined by titration in preliminary experiments with the target cells intended for the transfection, if necessary with a mixed cell population, and the vector system intended for the transfection, whereby a gene construct that is appropriate in terms of size is advantageously used as DNA largely corresponds to that intended for the specific application and which contains a reporter gene for the purpose of easier measurement of the efficiency of the gene transfer.
  • the suitability of the luciferase and the ⁇ -galactosidase gene as a reporter gene for such experiments has been demonstrated.
  • the invention relates to a method for introducing complexes of nucleic acid, a nucleic acid-binding substance and optionally an internalization factor, into higher eukaryotic cells.
  • the method is characterized in that the cells are brought into contact with an agent which has the ability to be taken up into the cells either on its own or as part of the nucleic acid complexes and the content of the endosomomes in which the nucleic acid Complexes localized after entering the cell are released into the cytoplasm.
  • simultaneous administration of the endosomolytic agent and nucleic acid complex is preferred, but the administration can also take place in succession, in the case of separate use the order is not critical as long as the administration is carried out in quick succession by one to ensure simultaneous possible contact of the cell with the components.
  • free virus is used in a separate preparation, the simultaneous administration of the virus preparation with the complexes can be ensured in that the virus preparation is part of the transfection medium which contains the nucleic acid complex.
  • the nucleic acid complexes and the virus preparation are mixed before use.
  • the endosomolytic agent is used as part of a combination complex.
  • compositions according to the invention can also be used repeatedly.
  • the cells are primary tumor cells.
  • the nucleic acid is a DNA which contains one or more sequences which code for an immunomodulating substance, preferably a cytokine.
  • the cells are myoblasts, preferably primary myoblasts.
  • the cells are fibroblasts, preferably primary fibroblasts.
  • the cells are hepatocytes, preferably primary hepatocytes. In another embodiment, the cells are primary endothelial cells.
  • the cells are primary airway epithelial cells.
  • the cells are T cells.
  • the cells are B cells.
  • Table 1 shows the successful transfection with the help of the present invention using the example of different cell types.
  • composition according to the invention was also examined in the transfection of canine hemophilia B fibroblasts. Both luciferase and ⁇ -galactosidase could be successfully expressed in these cases.
  • the system was also used to introduce the 1.4 canine factor IX cDNA into these fibroblasts. Using sandwich ELISA, factor IX could be detected 24 hours after the transfection.
  • a lysosomatropic substance in addition to the endosomolytic agent, e.g. if the endosomolytic agent is a peptide conjugate or a retrovirus, the endosomolytic activity of which is not strictly pH-dependent.
  • lysosomatropic substances inhibit the activity of proteases and nucleases and thus can inhibit the breakdown of nucleic acids (Luthmann and Magnusson, 1983). These substances include Chloroguin, Monensin, Nigericin and Methylamine. It has been shown that monensin causes an increase in reporter gene expression when Moloney virus is used. It could be shown that the presence of chloroguin in practically 100% of K562 cells leads to the expression of a reporter gene which was introduced into the cells by transferrin-mediated DNA transfer.
  • BNL.CL2 or HepG2 hepatocytes did not respond as well to chloroguin as the K562 cells, but could be transfected to a level of 5 to 10% when the endosomolytic properties of added replication-defective or chemically inactivated adenovirus were exploited.
  • the advantages of the biological vectors are enhanced. Because of the distribution of the receptors, there is a tropism for both the internalizing factor and the virus. By matching these two components to the respective cell population, an increased selectivity can be achieved, which is particularly important in the therapeutic application of the present invention.
  • This aspect is of particular importance in the therapeutic application of the present invention in the lungs because the different cell populations in the lungs have different receptors, which require the construction of vectors with high binding affinity for a specific cell population, for example for the ciliated cells of the respiratory tract can.
  • Lectins for example, are suitable as ligands.
  • the construction of such conjugates requires confirmation of the binding properties of a ligand candidate in the conjugate conformation. This confirmation can, for example, with antibodies against the Ligands can be carried out using immunohistochemical staining methods in the tissue in which the therapeutic application is to take place.
  • the present invention relates to pharmaceutical compositions which contain as an active ingredient a complex of therapeutically active nucleic acid, preferably as part of a gene construct, and an endosomolytic agent which is optionally conjugated and, if appropriate, an internalizing factor conjugate.
  • a complex of therapeutically active nucleic acid preferably as part of a gene construct
  • an endosomolytic agent which is optionally conjugated and, if appropriate, an internalizing factor conjugate.
  • Any inert pharmaceutically acceptable carrier can be used, e.g. Saline or phosphate buffered saline or any carrier in which the DNA complexes have suitable solubility properties to be used in the present invention.
  • the present invention offers the advantage of the greatest possible flexibility in use, inter alia as a pharmaceutical preparation.
  • the composition according to the invention can be present as a lyophilisate or in a suitable buffer in the deep-frozen state. It can also be provided as a ready-to-use reagent in solution, preferably transported and stored refrigerated. If necessary, the components required for the transfection, namely DNA, endosomolytic agent, optionally conjugated or ready for conjugation with a separate conjugation partner, DNA-binding substance, optionally conjugated with an internalization factor, optionally free polycation, are separated or partially separated in a suitable buffer as components of a Transfection kits, which is also the subject of the present invention.
  • the transfection kit of the present invention comprises a carrier which contains one or more containers, such as tubes, vials or the like, which contain the materials required for the transfection of the higher eukaryotic cell according to the present invention.
  • a first container can contain one or more different DNAs, for example coding for different antigens.
  • a second container can contain one or more different internalization factor conjugates, the use of the transfection kit in the form of a modular system being made possible. Whether the ingredients are available as a ready-to-use preparation or separately to be mixed immediately before use depends on the specific application and the stability of the complexes, which can be routinely examined in stability tests.
  • a transglutaminase-coupled adenovirus-polylysine conjugate which has been found to be stable in storage, is located in one of the containers of a kit.
  • biotinylated adenovirus and streptavidin polylysine are in separate containers and are mixed before use. Taking advantage of the flexibility of the invention, those of ordinary skill in the art can develop numerous different transfection kits.
  • composition according to the invention as a pharmaceutical preparation can take place systemically, the intravenous route being preferred.
  • Target organs for this type of application are, for example, the liver, spleen, lungs, bone marrow and tumors.
  • Examples of local application are the lung tissue (application of the composition according to the invention as a liquid for instillation or as an aerosol for inhalation).
  • lung tissue application of the composition according to the invention as a liquid for instillation or as an aerosol for inhalation.
  • the pharmaceutical preparations according to the invention can be administered by direct injection into the liver, into muscle tissue or into a tumor or by local application in the gastrointestinal cycle.
  • Another method for the administration of the pharmaceutical composition is application via the bile duct system. This method of application allows direct access to the hepatocyte membranes on the biliary tubules, avoiding the interaction of the composition with blood components.
  • myoblasts implant muscle cells
  • myoblasts are small muscle cells
  • This method can be used more widely than for the treatment of genetic defects in muscle cells, such as the defect related to muscular dystrophy.
  • the manipulated myoblasts can thus be used to deliver gene products that either act in the blood or are transported by the blood.
  • the experiments of the present invention have shown that both myoblast and myotube cultures, even primary ones, can be transfected with high efficiency.
  • the Transfection media that showed the best results contained combination complexes of biotinylated adenovirus, transferrin-polylysine and streptavidin-polylysine.
  • factor VIII was expressed in the muscle cells.
  • the chicken adenovirus CELO was used in combination complexes containing wheat germ agglutinin as an additional internalizing factor.
  • the therapeutic application can also take place ex vivo, the treated cells, e.g. Bone marrow cells, hepatocytes or myoblasts are returned to the body, e.g. Ponder et al., 1991, Dhawan et al., 1991.
  • Another ex vivo application of the present invention relates to so-called cancer vaccines.
  • the principle of this therapeutic approach is to isolate tumor cells from a patient and to transfect the cells with a DNA coding for a cytokine.
  • the cells are inactivated if necessary, e.g. by radiation in such a way that they no longer reproduce but still express the cytokine.
  • the genetically modified cells are then administered to the patient from whom they were taken as vaccine.
  • the secreted cytokines activate the immune system, including by activating cytotoxic T cells. These activated cells can act in other parts of the body and also attack untreated tumor cells. This reduces the risk of tumor recurrence and metastasis development.
  • a suitable protocol for the use of cancer vaccines for gene therapy was described by Rosenberg et al., 1992. Instead of the retroviral vectors proposed by Rosenberg, the gene transfer system of
  • REPLACEMENT LEAF present invention can be used.
  • primary melanoma cells were successfully transfected with a reporter gene contained in combination complexes of polylysine-coupled adenovirus and transferrin-polylysine or LDL-polylysine.
  • the present invention can also be used in assays to determine the immune response to a given antigen.
  • Antigen-specific cytotoxic T-lymphocytes CTL
  • CTL cytotoxic T-lymphocytes
  • APC antigen presenting cells
  • HLA Human Lyphocytic Antigens
  • MHC MHC, “Major Histocompatibility Molecules”
  • CTL Killing Assay the antigen must be presented to the CTLs in the correct HLA context, which usually means on the autologous target cell.
  • a "CTL killing assay” can be carried out as follows: APCs are transfected with a DNA construct which contains a sequence coding for an antigen. Antigen epitopes are bound to MHC class I molecules and presented on the cell surface as a target for a specific CTL response. After incubation with a sample of patient serum, the APCs are lysed depending on the presence of specific CTLs. Cell lysis can be measured, for example, by monitoring the release of radioactive chromium that was introduced into the APCs before the serum was added.
  • B-LCLs B-lymphoblastoid cell lines
  • CTL Killing Assays use the gene transfer system of the present invention to introduce DNA encoding antigens into the cell, for example to insert constructs encoding HIV or tumor antigens into fibroblasts to do this To have antigen expressed.
  • Primary fibroblasts can easily be obtained from biopsies, are easy to grow and have proven to be particularly efficient (approx. 50 to approx.
  • transfectable with the aid of the present invention Such assays are useful in identifying epitopes that are recognized by killer cells in view of vaccine development. They can also advantageously be used to determine a person's HLA-restricted immune response to a particular antigen.
  • the invention can also be used to produce recombinant proteins.
  • the invention can also be used to produce recombinant proteins.
  • Almost any cell type can therefore be used for the production of recombinant proteins, which ensures that the recombinant protein is produced in a true-to-nature and in a completely modified post-translationally processed form, which ensures the high biological activity of the product.
  • the gene transfer into the cells can be achieved, as shown in the present examples with the aid of luciferase and IFN- ⁇ , practically any gene construct which leads to the formation of a desired protein product can be transported.
  • the desired protein product can be obtained from the cell culture 24 hours to 1 week or longer after the transfection (either from the cell supernatant or from a suitable cell homogenate, in accordance with the instructions for the respective protein product).
  • Gene expression can take place in cells which ensure that the corresponding post-translational processing and modification takes place (e.g. vitamin K-dependent carboxylation of coagulation factors, see Armentano et al., 1990, or cell-type-specific glycosylation.
  • post-translational processing and modification e.g. vitamin K-dependent carboxylation of coagulation factors, see Armentano et al., 1990, or cell-type-specific glycosylation.
  • the system according to the invention makes a larger selection of target cells available for gene expression.
  • FIG. 1 Effect of adenovirus infection on the
  • Fig. 2 Conjugate-DNA complex dose effect
  • Fig. 3 Enhancement of transferrin-polylysine-mediated gene transfer by adenoviruses takes place via receptor-mediated endocytosis:
  • Fig. 4 Effect of adenovirus infection on the
  • Fig. 12 Inactivation of adenoviruses by formaldehyde
  • HE Fig. 13 Transfection of NIH3T3 cells with
  • FIG. 14 Investigation as to whether the gene transfer effect in the transfection of NIH3T3 cells with transferrin-polylysine-DNA complexes is due to Moloney virus.
  • FIG. 15 Interactions between transrin and its receptor play a role in the gene transfer effect of Moloney virus.
  • FIG. 16 Influence of the pH on the gene transfer effect of retroviruses
  • FIG. 17 Influenza hemagglutinin peptide; Liposome
  • Fig. 18 Transfection of K562 cells with transferrin-polylysine conjugates in the presence of influenza peptide-polylysine conjugate
  • Fig. 19 Transfection of HeLa cells with transferrin-polylysine conjugates in the presence of influenza peptide-polylysine conjugate
  • Fig 20 In situ detection of the ⁇ -galactosidase
  • Fig. 21 In situ ß-galactosidase expression in HeLa cells in the presence of adenovirus
  • Fig. 22 Transfection of cells with a 48 kb cosmid in the presence of adenovirus.
  • A Heia cells.
  • B Neuroblastoma cells
  • Fig. 23 Production of adenovirus polylysine by chemical coupling
  • Fig. 24 Transfection of K562 cells with chemically coupled adenovirus conjugates
  • Fig. 25 Transfection of HeLa cells with chemically coupled adenovirus conjugates
  • Fig. 26 Binding of polylysine to adenovirus by means of Transglutaminase
  • Fig. 27 Transfection of mouse hepatocytes using transglutaminase-coupled adenovirus-polylysine conjugates
  • Fig. 31 Transfection of neuroblastoma cells with a 48 kb cosmid using biotin-streptavidin-coupled adenovirus conjugates
  • Fig. 32 Transfection of hepatocytes in the presence of chloroguin or adenovirus
  • Fig. 33 Transfection of K562 cells in the presence of various endosomolytic agents
  • Fig. 34 Comparison of the transfection protocols at the cellular level with ⁇ -galactosidase as reporter gene in the presence of various endosomolytic agents
  • Fig. 35 Long-term luciferase expression in confluent, non-dividing hepatocytes
  • Fig. 38 Transfection of primary myoblast and myotube cultures
  • FIG. 39 Comparison of adenovirus dl312 and CELO virus in the transfection of HeLa cells and C2C12 myoblasts.
  • FIG. 40 improvement of the transfection with CELO virus by using a lectin ligand
  • FIG. 41 expression of a factor VIII cDNA in C2C12
  • Fig. 42 Enhancement of DNA transport by adenovirus proteins.
  • A HeLa cells.
  • B Fibroblasts.
  • Fig. 43 Galactose-influenza apeptide conjugates for the
  • Fig. 45 Gene transfer in B lymphoblastoid B cells
  • Fig. 46 DNA transfer with transferrin polylysine in the presence of rhinovirus.
  • A free rhinovirus.
  • B conjugated rhinovirus
  • Fig. 47 transfection of primary human
  • Fig. 49 Gene transfer in respiratory epithelial cells from
  • Fig. 52 Transfection of BNL CL.2 cells in the presence of amphipathic peptides
  • Fig. 53 Transfection of NIH3T cells in the presence of amphipathic peptides
  • Fig. 54 Expression of IFN- ⁇ in HeLa cells, transfected in the presence of various endosomolytic agents
  • the modified transferrin solution was rapidly (within 10 to 15 min) a solution containing 1.5 ⁇ mol fluorescent-labeled poly (L) lysine with a average chain length of 190 lysine monomers in 4.5 ml of 100 mM sodium acetate pH 5 added. The pH of the solution was adjusted to pH 7.5 by adding 1 M sodium bicarbonate buffer.
  • the transferrin conjugates if they were not used immediately, were shock-frozen in liquid nitrogen at -20 ° C in an iron-free form. Before iron was incorporated, samples (0.5 to 1 mg) were brought to a physiological salt concentration (150 mM) with sodium chloride. Iron incorporation was accomplished by adding 4 ul 10mM iron (III) citrate buffer (containing 200mM citrate)
  • the plasmid pCMV was prepared by removing the BamHI insert of the plasmid pSTCX556 (Severne et al., 1988), treating the plasmid with Klenow fragment and the HindIII / Sspl and Klenow-treated fragment from the plasmid pRSVL, which is the one for luciferase contains coding sequence, or which was used for ⁇ -galactosidase (Macgregor and Caskey, 1989).
  • the complex formation was carried out analogously to pRSVL.
  • storage buffer 100 mM Tris, pH 8.0, 100 mM NaCl, 0.1% BSA, 50% glycerol
  • the virion concentration was determined by means of UV spectrophotometric analysis of the extracted genomic virus DNA (formula: an optical density unit (OD, A260) corresponds to 10 - * - 2 virus particles / ml; (Chardonnet and Dales, 1970)).
  • the Moloney mouse leukemia retrovirus N2 was packaged in an ecotropic packaging line (Keller et al., 1985, Armentano et al., 1987). Supernatants expressing viral cell lines were collected, flash frozen in liquid nitrogen and stored at -20 C ⁇ . The supernatants used in the examples had a titer of approximately 106 cfu / ml, which was measured by neomycin resistance colonization with NIH3T3 cells. For the virus concentration experiments, the supernatants were passed through a 300 Kb membrane filter (FILTRON) in an AMICON stirred cell concentrator under nitrogen pressure. Normally 10 - 30 ml of supernatant could be concentrated tenfold.
  • FILTRON 300 Kb membrane filter
  • HeLa cells were grown in DMEM medium supplemented with 5% heat inactivated fetal calf serum (FCS), penicillin at 100 IU / ml, streptomycin at 100 ⁇ g / ml and 2 mM glutamine.
  • FCS heat inactivated fetal calf serum
  • WI-38, MRC-5, and KB cells were grown in EMEM medium (Eagle's modified essential medium) supplemented with 10% heat-inactivated FCS, antibiotics such as DMEM medium, 10 mM non-essential amino acids and 2 mM glutamine.
  • CFT1 a respiratory cystic fibrous epithelial cell line (made according to the method described by Yankaskas et al., 1991; the CFT1 cell line is characterized by being homozygous for the ⁇ F508 deletion CF mutation) was found in F12-7X Medium grown (Willumsen et al., 1989). For the gene transfer experiments, the cells were grown in 6 cm cell culture plates until they were approximately 50% confluent (5 ⁇ 105 cells). The medium was removed and 1 ml DMEM or EMEM / 2% FCS medium added.
  • the conjugate-DNA complexes were then added, followed immediately by the adenovirus dl312 (0.05 - 3.2 x 104 particles / cell) or a comparable volume of virus storage buffer (1 - 80 ⁇ l).
  • the plates were returned to the incubator for one hour (5% CO 2, 37 ° C), then 3 ml of complete medium was added. After another 24 h of incubation, the cells were harvested to determine luciferase gene expression. In the case of the CFTI cells, the cells were grown for 4 hours in F12-7X medium without human transferrin before the gene transfer experiments.
  • HeLa cells CCL 2
  • K562 cells CCL 243
  • HepG2 cells HB 8065
  • TIB-73 cells TIB 73 (BNL CL. 2)
  • NIH3T3 cells CRL 1658
  • 293 cells CRL 1573
  • KB cells CCL 17, WI-38 cells: CCL 75
  • MRC-5 cells CCL 171.
  • H9 cells were obtained from AIDS Research and Reference Reagent Program, US Department of Health and Human Services, catalog no. 87.
  • Primary lymphocytes were obtained by taking a 25 ml sample of umbilical cord blood in test tubes containing EDTA. Aliquots were placed under 4.5 ml of Ficoll-hypague (Pharmacia) and centrifuged for 15 minutes at 2500 rpm. The brownish layer between the upper plasma layer and the clear Ficoll layer was removed (approx. 10 ml). 40 ml of IMDM plus 10% FCS were added, the sample was centrifuged at 1200 rpm for 15 min and the cell pellet was taken up in 50 ml of fresh IMDM plus 10% FCS (the cell density was approx. 2 ⁇ 106 cells / ml).
  • the cells were divided 1: 3 with IMDM / 20% FCS, 2 units / ml IL-2. Aliquots of the cells were frozen in liquid nitrogen in FCS plus 5% DMSO. Before use, the cells were grown in IMDM plus 20% FCS plus 2 units / ml IL-2.
  • HeLa cells were equilibrated at 4 ° C in 1 ml DMEM, supplemented with 2% FCS.
  • the conjugate-DNA complexes were added as in the other experiments and the plates were incubated for 2 hours at 4 ° C.
  • the plates were then washed extensively with ice-cold DMEM / 2% FCS, then 2 ml of this medium were added.
  • Adenovirus dl312 or virus buffer was then added and the cells were allowed to warm up slowly before being placed in the incubator for a further 24 h. After this incubation, the cells were harvested and examined for luciferase gene expression.
  • Luciferase activity was carried out as described by Zenke et al., 1990, Cotten et al., 1990, or in the
  • Adenoviruses enhance the gene transfer caused by transferrin-polylysine via receptor-mediated endocytosis a) Effect of adenovirus treatment on the transfer of complexed DNA
  • Conjugate-DNA complexes (DNA + hTfpL190B) or polylysine-DNA complexes (DNA + pL) were bound to HeLa cells without being internalized by incubating at 4 ° C. Unbound complex was removed prior to adding adenovirus dl312 (1 x 104 virus particles / cell) or a comparable buffer volume. The subsequent incubation was carried out at 37 ° C. to allow internalization of the bound DNA complexes and the adenoviruses. The luciferase activity was determined as indicated (FIG. 3B).
  • Conjugate-DNA complexes containing 6 ⁇ g pRSVL-DNA plus 12 ⁇ g transferrin-polylysine (DNA + hTfpL190B) were added to the HeLa cells at a concentration of 1 ⁇ 10 4 adenovirus particles (dl312) / cell or a comparable amount of heat-inactivated adenovirus dl312 (dl312 hi). Heat inactivation was carried out by incubation at 45 ° C. for 30 min (Defer et al., 1990).
  • Conjugate-DNA complexes (6 ⁇ g pRSVL + 12 ⁇ g hTfpL190B) were added to cells of the cell lines CFT1, KB, HeLa, WI38 and MRC5 with or without adenovirus dl312 (1 ⁇ 10 4 virus particles / cell). The efficiency of gene transfer for the various cell lines was determined by means of the luciferase assay as in the previous examples (FIG. 4).
  • a cell line constitutively expressing luciferase called K562 10/6 was first produced by transfecting cells with a plasmid that contains an RSV luciferase gene fragment (an Apal / Pvul fragment from pRSVL (De Wet et al., 1987)) , cloned into the clal site of the pUC ⁇ locus (Collis et al., 1990).
  • This plasmid was complexed with a transferrin-polylysine conjugate and K562 cells were transfected with these complexes, following the method described by Cotten et al., 1990.
  • pUC ⁇ locus plasmid contains a neomycin resistance gene
  • clones expressing luciferase could be selected due to the neomycin resistance.
  • a clone called K562 10/6 was selected for the further experiments.
  • Aliguots of the parental cell line K562 (in 200 ⁇ l RPMI 1640 plus 2% FCS; 500,000 cells / sample) were treated either with 12 ⁇ g TfpL plus 6 ⁇ g pRSVL or with 4 ⁇ g pL90 plus 6 ⁇ g pRSVL, each in 500 ⁇ l HBS.
  • the indicated amounts of adenovirus dl312 (Fig. 5) were allowed to act on the cells at 37 ° C for 1.5 hours, after which 2 ml of RPMI and 10% FCS were added. The incubation was then continued at 37 ° C. for a further 24 h and the cells were then prepared for the determination of luciferase activity.
  • luciferase activity (Fig. 5A). This applies to both the TfpL complexes (200 light units versus 25,000 light units) and the pL90 complexes (0 versus 1.9 x 106 light units).
  • the K562 cell line has the ability to internalize pRSVL / polylysine complexes and that this internalization, as measured by luciferase expression, is increased significantly by the presence of adenovirus.
  • liver cells with asialofetuin-polylysine conjugates (AFpL) or with tetra-galactose peptide-pL conjugates ((gal) 4pL) in the presence of adenovirus
  • Conjugates were prepared by mixing 1.5 ⁇ mol of the lactosylated peptide prepared in a) in 3 ml of 20 mM NaCl with 0.146 ⁇ mol of the modified pL290 obtained from b) in 620 ⁇ l of 100 mM sodium acetate buffer under an argon atmosphere. After adding 100 ⁇ l
  • conjugates were prepared on the same principle as the transferrin conjugates; a similar procedure for the preparation of asialoorosomucoid-polylysine conjugates has been described by Wu and Wu, 1988.
  • Conjugates were prepared by mixing 1.4 ⁇ mol of modified asialofetuin in 5 ml of 100 mM HEPES pH 7.9 with 0.33 ⁇ mol of modified pL190 (containing 1.07 ⁇ mol of mercaptopropionate groups; the same procedure was followed for the preparation of the transrin conjugates) in 6.5 ml of 200 mM HEPES pH 7.6 were mixed under an argon atmosphere. The reaction mixture was left to stand at room temperature for 24 hours.
  • the conjugates were removed from the reaction mixture by cation exchange chromatography (Pharmacia Mono S-pillar HR 10/10; Gradient elution, buffer A: 50 mM HEPES pH 7.9; Buffer B: Buffer A plus 3 M sodium chloride) isolated and sodium chloride added to a final concentration of 0.6 M before loading the column.
  • the product fraction eluted at a salt concentration of approx. 1.5 M.
  • Dialysis against HBS gave conjugates containing 0.52 ⁇ mol asialofetuin, modified with 0.24 ⁇ mol pL190.
  • HepG2 cells were grown in DMEM medium plus 10% FCS 100 IU / ml penicillin, 100 ⁇ g / ml streptomycin and 2 mM glutamine in T25 bottles. The transfections were carried out at a density of 400,000 cells / bottle. Before transfection, the cells were washed with 4 ml of fresh medium containing 10% FCS. Chloroguin (Sigma) was added immediately before the transfection, so that the final concentration in the cell suspension (plus DNA solution) was 100 ⁇ M.
  • the mixture was added to the cells; the cells were incubated for 4 hours at 37 ° C, then the transfection medium was replaced with 4 ml of fresh DMEM medium plus 10% FCS. After 24 hours, the cells were harvested for the luciferase assay.
  • the values shown in Fig. 7 represent the
  • HepG2 cells were grown in 6 cm plates to a cell density of 300,000 cells / plate as indicated in e). Before transfection, the cells were washed with 1 ml of fresh medium containing 2% FCS.
  • lactosylated peptide (gal) 4pL ((gal) 4pLl + (gal) 4 or (gal) 4pL2 + (gal) 4) were added. The mixture was added to the cells; the cells were
  • Cells of the embryonic mouse liver cell line ATCC TIB73 (BNL CL.2; Patek et al., 1978) were DMEM (0.4% glucose) supplemented with 10% heat-inactivated FCS at 37 ° C. in a 5% CO 2 atmosphere in "high glucose" , containing 100 IU / ml penicillin, 100 ⁇ g / ml streptomycin and 2 mM glutamine, grown in 6 cm plates.
  • the transfections were carried out at a cell density of 300,000 cells / plate. Before transfection, the cells were washed with 1 ml of fresh medium plus 2% FCS.
  • a transfection without adenovirus was carried out in the presence of chloroguin: the Trans ections were carried out at a cell density of 300,000 cells / plate. Before transfection, the cells were washed with 1 ml of fresh medium containing 2% FCS. Chloroguin (Sigma) was added immediately before the transfection, so that the final concentrations in the cell suspension (plus DNA solution) were 100 ⁇ M. 6 ⁇ g pCMVL-DNA in 330 ⁇ l HBS were mixed with the stated amounts of mTfpL, AFpL, pLys290, (gal) 4pLl or (gal) 4pL2 in 170 ⁇ l HBS.
  • the DNA complexes were added to the cells.
  • the cells were incubated at 37 ° C for 2 hours, then 1.5 ml of medium containing 10% FCS and 100 ⁇ M chloroquine were added. Two hours later, the transfection medium was replaced with 4 ml of fresh medium. After 24 hours, the cells were harvested for luciferase determination. The values obtained for the luciferase activity are shown in Fig. 9B.
  • a solution of 1.3 mg of antiCD7 antibody (Immunotech) in 50 mM HEPES pH 7.9 was mixed with 49 ⁇ l of 1 mM ethanolic solution from SPDP (Pharmacia). After 1 h at room temperature, the mixture was filtered through a Sephadex G-25 gel column (eluent 50 mM HEPES buffer pH 7.9) to give 1.19 mg (7.5 nmol) antiCD7, modified with 33 nmol pyridyldithiopropionate residues.
  • Poly (L) lysine90, fluorescent labeling using FITC was modified analogously with SPDP and brought into the form modified with free mercapto groups by treatment with dithiothreitol and subsequent gel filtration.
  • a solution of 11 nmol polylysine90, modified with 35 nmol mercapto groups, in 0.2 ml 30 mM sodium acetate buffer was mixed with modified antiCD7 (in 0.5 ml 300mM HEPES pH 7.9) in the absence of oxygen and left overnight at room temperature.
  • the reaction mixture was brought to a level of approximately 0.6 M by adding 5 M NaCl.
  • the conjugates were isolated by ion exchange chromatography (Mono S, Pharmacia, 50 mM HEPES pH 7.3, salt gradient 0.6 M to 3 M NaCl); after dialysis against 10 mM HEPES pH 7.3, appropriate conjugates consisting of 0.51 mg (3.2 nmol) antiCD7 antibody were modified with
  • conjugates were isolated by ion exchange chromatography (Mono S, Pharmacia, 50 mM HEPES pH 7.3, salt gradient 0.6 M to 3 M NaCl); after fractionation and dialysis against 25 mM HEPES pH 7.3, three conjugate fractions A, B and C were obtained, consisting of 0.40 mg rgpl20 modified with 1.9 nmol polylysine 190 (in the case of fraction A) or 0.25 mg rgp 120 modified with 2.5 nmol polylysine 190 (fraction B), or 0.1 mg rgp 120 modified with 1.6 nmol polylysine 190 (fraction C).
  • pCMVL-DNA (6 ⁇ g / sample) was complexed with the specified amounts of Polylysin90 or the specified Polylysin conjugates in 500 ⁇ l HBS.
  • H9 cells 106 cells in 5 ml RPMI with 2% FCS
  • primary human lymphocytes 3 x 106 cells in Iscove's modified Dulbecco's medium (IMDM) plus 2% FCS
  • IMDM Iscove's modified Dulbecco's medium
  • FIG. 10A H9 cells
  • FIG. 10B primary Lymphocytes
  • adenovirus dl312 preparation prepared and stored as described in the introduction to the examples, was placed in the 2 cm wells of a cell culture plate (300 ⁇ l / well) on ice at 8 cm distance from two UV lamps (Philips TUV15 (G15 T8 ) - lamps). The virus was exposed to UV radiation for the duration of the times shown in FIG. 11A and aliguots of each preparation were examined for their virus titer and for their ability to determine whether and to what extent they are capable of gene transfer with polylysine-transferrin conjugates in HeLa cells.
  • Fig. 11A The complexes from pCMVL-DNA and 12 ⁇ g TfpL were prepared in 500 ⁇ l HBS and 3 ⁇ 105 HeLa cells (in 1 ml DMEM plus 2% FCS) were added. Approximately 5 min later, 54 ul of each virus preparation of each culture was added and the culture incubated at 37 ° C for one and a half to two hours. A 5 ml aliquot of DMEM plus 10% FCS was then added to each culture, incubation continued at 37 ° C for 24 hours and the cultures harvested for luciferase activity examined.
  • the amount of 54 ⁇ l of non-irradiated virus is not in the saturation range, ie the test is sensitive to at least 3 times the amount of virus.
  • the values obtained for the luciferase expression are shown in FIG. 11B (closed squares).
  • the virus titer of each preparation was determined using the E1A complementing cell line 293. Serial dilutions of the unirradiated and irradiated virus samples were first prepared in DMEM plus 2% FCS. In parallel, samples of 5 x 10 4 293 cells (in a 2 cm well) were prepared and placed in 200 ⁇ l DMEM plus 2% FCS. A 5 ul aliquot of each dilution was placed in a second well for comparison. To effect virus binding to the cells, incubation was carried out at 37 ° C for one and a half hours, then 2 ml of DMEM plus 10% FCS was added to each well. The cultures were counted 48 hours later to determine the cytopathic effect.
  • the virus dilution above which less than 50% of the cells in the culture show a clear cytopathic effect after 48 h indicates the relative amount of infectious virus in each virus preparation.
  • the values obtained are shown in Fig. 11B (open squares).
  • the result of the experiments carried out in this example shows that a decrease by 4 orders of magnitude in the virus titer by UV irradiation brings about only a 20-fold reduction in the luciferase gene transfer. This shows that mechanisms that govern the infectivity of the virus can be destroyed without affecting the ability of the virus to enhance gene transfer.
  • adenovirus preparation 2 ml of adenovirus preparation were passed through a 10 ml G25 column (Pharmacia PD 10 G, 25 M), pre-equilibrated with 150 mM NaCl, 25 mM HEPES pH 7.9, 10% glycerol and collected in a volume of 2.5 ml. Aliguots of the gel-filtered virus preparation were incubated on ice without (0), with 0.01%, 0.1% or 1% formaldehyde for 20 h.
  • Tris pH 7.4 was added to a concentration of 100 mM, then the samples were first against 1 1 150 mM NaCl, 50 mM Tris pH 7.4 and 50% glycerol and then overnight against 2 x 1 1 150 mM NaCl, 20 mM HEPES pH 7.9 and 50% glycerin dialyzed.
  • the virus preparations were examined on the one hand for their ability to enhance gene transfer by means of pCMVL / hTfpL complexes in HeLa cells (represented as light units, left axis in FIG. 12B), and on the other hand for their ability to replicate in 293 cells (virus titers , left axis in Fig. 12B).
  • Transferrin-polylysine-90 conjugates and conjugate-DNA complexes were prepared analogously to the preceding examples, with the difference that the complex formation reaction was carried out in a volume of 500 ⁇ l mM NaCl, 20 mM HEPES, pH 7.4.
  • NIH3T3 cells were grown in DMEM medium with the addition of 10% FCS, 100 IU / ml penicillin, 100 ⁇ g / ml streptomycin and 2 mM glutamine.
  • 5-7 x 105 cells per T25 bottle were plated 18 to 24 hours before the transfection.
  • the cells were placed in fresh medium and the various components used for the transfection were added in the following order: chloroquine (100 ⁇ M, where indicated), polylysine-transferrin-DNA complex, retrovirus preparation.
  • the cells were then incubated at 37 ° C for 4 hours, then the medium was changed and the cells were harvested 24 hours later. Extracts were made using three freeze / thaw cycles; Aliguots of the extract, standardized for protein content, were tested for luciferase activity, as indicated for the previous examples.
  • transfections of 106 NIH3T3 cells with TfpL-DNA complexes were carried out in the presence of 100 ⁇ M chloroquine or without chloroquine, as indicated in FIG. 13. It was found that without chloroguin, the values for the luciferase activity only reached background levels (column 1), while in the presence of chloroquine a high expression of the pRSVL reporter gene was measured
  • REPLACEMENT LEAF could (column 2). Increasing amounts of the Moloney leukemia virus, which were added to the cells at the same time as the DNA complexes, could cause an increasing increase in the luciferase gene expression. (The amounts shown in Fig. 13 are ml.)
  • the virus preparation used in Example 9 was a crude, unfractionated supernatant from cells expressing retrovirus.
  • the supernatant was subjected to the dialysis / concentration purification described above, the retrovirus supernatant (in Fig. indicated with RVS) was concentrated by a factor of 10. If the retrovirus is responsible for the amplification, the activity found in the membrane residue should be about 10 times the original excess, apart from any inactivation of the extremely labile retrovirus during the concentration step.
  • 106 NIH3T3 cells were transfected under the conditions shown in FIG. 14.
  • the retrovirus was examined for its ability to insert plasmid DNA into the cell transport that is complexed only with polylysine.
  • the amount of polylysine used corresponds to the previously determined optimum, which brings about complete condensation of the plasmid DNA and is similar to the amount of polylysine which is used with the polylysine-transferrin conjugate (Wagner et al., 1991a). The experiments, the result of which is shown in FIG.
  • the peptide of the sequence (SEQ ID N0: 1) Gly-Leu-Phe-Glu-Ala-Ile-Ala-Gly-Phe-Ile-Glu-Asn-Gly-Trp-Glu-Gly-Met-Ile-Asp-Gly -Gly-Gly-Cys was synthesized using the Fmoc (fluorenylmethoxycarbon 1) method (Aether on et al., 1979) using an Applied Biosystems 431A peptide synthesizer.
  • the side chain protecting groups were t-butyl for Cys, Glu and Asp and trityl for Asn.
  • the mixture was then dialyzed twice against 2120 mM HEPES / 0.5 M guanidine hydrochloride.
  • the solution obtained was applied to a Mono S column (0.7 x 6 cm, Pharmacia) (gradient: 0-20 min 100% A, 20-140 min 0-100% B.
  • the product fraction which eluted with 1.5 M guanidine hydrochloride, was dialyzed against 2 ⁇ 2 1 HBS.
  • the subsequent determination of the pL concentration by the ninhydrin test showed a concentration of approximately 1.14 mg / ml.
  • the amount of peptide in the solution of the conjugate was calculated from its absorption at 280 nm; this resulted in a molar ratio of peptide: L of 4: 1.
  • thiopyridone After reducing an aliquot of the filtrate with DTT, a determination of thiopyridone was carried out, which showed a content of 3.62 ⁇ mol SPDP in the product fraction.
  • the SPDP-modified pL was obtained by adding 79 mg of DTT reduced to the solution. After a 2 h reduction, the solution was filtered again on G25 under the specified conditions. The thiol determination using the Ellmann test showed a thiol concentration of 3.15 ⁇ mol in 2,224 ml.
  • Liposomes were produced using the REV method ("reverse phase evaporation") (Szoka and Papahadjopoulos, 1978; Straubinger and Papahadjopoulos, 1983): aqueous phase 10 mM HEPES pH 7: 3, 100 mM calcein 150 mM NaCl; organic phase: a solution of 300 ⁇ mol L- ⁇ -lecithin (from egg yolk, mainly palmitoyloleoylphosphatidylcholine; Avanti Polar Lipids) in 260 ⁇ l chloroform was evaporated using a rotary evaporator. The material was then dried in a high vacuum and then redissolved in 3 ml of diethyl ether.
  • REV method reverse phase evaporation
  • the resulting emulsion (1.7 ml) was centrifuged at 500 rpm and then extruded through a Nucleopore polycarbonate membrane (0.1 ⁇ m), resulting in a final volume of 0.7 ml of liposome solution.
  • the liposomes were separated from the non-incorporated material by gel filtration (Sephadex G-50 medium, Pharmacia; 23 ml gel volume, 10 mM HEPES pH 7.3 / 150 mM NaCl).
  • Six 500 ⁇ l fractions were collected. Lipid phosphorus was determined to be 2 mM by the Bartlett method, 1959.
  • the release of the liposome content (“leakage”) was measured on the basis of the leakage of the enclosed calcein and the resulting dilution, which causes the self-quenching of the fluorescence to be abolished (Bondeson et al., 1984).
  • the calcein fluorescence was measured with a Kontron SMF 25 spectral fluorimeter (excitation at 490 nm, emission at 515 nm).
  • K562 cells were suspended in RPMI 1640 medium (Gibco BRL plus 2 g sodium bicarbonate / 1) plus 10% FCS, 100 units / ml penicillin, 100 ⁇ l / ⁇ l streptomycin and 2 mM glutamine to a density of 500,000 cells / ml bred. 12 to 20 h before the transfection, the cells were placed in fresh medium containing 50 ⁇ M desferrioxamine (this measure was taken in order to achieve an increase in the number of transferrin receptors). On the morning of the transfection, the cells were collected, suspended in fresh medium containing 10% FCS plus 50 ⁇ M desferrioxamine (250,000 cells / ml) and placed in a 24-well dish in each case 2 ml.
  • HeLa cells were grown in 6 cm culture dishes as indicated under "Cells and Media”. The transfections were carried out at a density of 300,000 cells / plate. Before transfection, the cells were incubated with 1 ml of fresh medium containing 2% FCS.
  • the peptide of the sequence (SEQ ID NO: 2) Gly-Leu-Phe-Gly-Ala-Ile-Ala-Gly-Phe-Ile-Glu-Asn-Gly-Trp-Glu-Gly-Met-Ile-Asp-Gly -Gly-Gly-Cys of the designation P41) was synthesized in an analogous manner to the peptide described in Example 13 a).
  • the coupling of the peptide to polylysine (pL300) was carried out as in Example 13 bl) by binding via SPDP. Conjugates with a molar ratio of peptide: polylysine of 4: 1 were obtained.
  • HeLa cells were grown in 6 cm dishes as indicated, the transfections were carried out at a density of 300,000 cells / dish. Before transfection, the cells were incubated with 1.5 ml of fresh medium containing 2% FCS. 6 ⁇ g pCMVL-DNA in 160 ⁇ l HBS (150 mM NaCl, 20 mM HEPES pH 7.3) were mixed with 6 ⁇ g of the conjugate TfpL190B in 160 ⁇ l HBS, after 15 min 10 ⁇ g influenza apeptide-polylysine conjugate P41pL or, for comparison, 18 ⁇ g of influenza peptide-polylysine conjugate P16pL (see example 13) added (FIG.
  • BNL CL.2 cells were grown as described in Example 6.
  • the influenza peptide P41 was conjugated to polylysine 300 at peptide: polylysine molar ratios of 1: 1, 3: 1 and 8: 1.
  • Complexes of 6 ⁇ g pCMVL-DNA and 20 ⁇ g of the conjugates were added to the cells.
  • 20 ⁇ g pL300 or 20 ⁇ g P16-polylysine conjugates, prepared as described in Example 13 were used.
  • the cells were incubated at 37 ° C for 4 hours, then 2 ml of medium containing 18% FCS was added. After 24 hours, the cells were harvested for the luciferase test, the results of which are shown in Figure 20B.
  • chloroquine was additionally added to the medium containing the DNA-polycation complexes at a final concentration of 100 ⁇ M or 50 ⁇ l of the adenovirus stock solution dl312C.
  • the original culture medium was removed from the cells and 1 ml of medium containing the DNA complexes with or without chloroquine or virus was added. After an incubation period of 2 hours at 37 ° C., 1 ml of DMEM, containing 10% FCS, antibiotics and glutamine, was added to the cells and the incubation was continued for a further 2 hours. Then all of the medium was removed and the cells were grown in 3 ml of fresh DMEM plus 10% FCS, antibiotics and glutamine.
  • the medium was removed 48 h after the transfection and the cells were buffered 1 ⁇ with phosphate Saline (PBS) washed and fixed with 0.5% glutardialdehyde in PBS for 5 min at room temperature. The fixative was then removed and the cells washed 1 ⁇ with PBS.
  • the staining solution (10 mM phosphate buffer pH 7.0, 150 mM NaCl, 1 mM MgCl2, 3.3 mM K4Fe (CN) 63H20, 3.3 mM K3Fe (CN) 6 and 0.2% 5-bromo-4-chloro-3-indolyl- ⁇ -galactopyranoside) at 37 "C for 20 min to 3 h (Lim and Chae, 1989).
  • the coverslips were then rinsed in PBS, water and 96% ethanol, dried and covered in Mowiol on slides.
  • a Zeiss Axiophot was used for analysis Microscope used.
  • FIG. 21 shows images of the microscopic magnifications (112 ⁇ ).
  • A HeLa cells, transfected with 6 ⁇ g pCMV-ß-gal, complexed with 12 ⁇ g TfpL190B. The staining reaction for ⁇ -galactosidase was carried out for 3 hours. The figure shows that very few cells (55 cells; the group of stained cells is indicated by an arrow) express the ⁇ -galactosidase gene.
  • B HeLa cells, transfected with 6 ⁇ g pCMV-ß-gal, complexed with 6 ⁇ g TfpL190B and 12 ⁇ g Pl ⁇ pL. Staining reaction: 3 h. Few cells (250 cells) express the ⁇ -galactosidase gene.
  • C HeLa cells, transfected with 6 ⁇ g pCMV-ß-gal, complexed with 6 ⁇ g TfpL190B and 12 ⁇ g P16pL in the presence of 100 ⁇ M chloroguin. Staining reaction: 3 h. Many groups of cells show a strong positive response (more than 1,000 cells).
  • D HeLa cells transfected with 6 ⁇ g pCMV-ß-gal, complexed with 12 ⁇ g TfpL190B in the presence of adenovirus dl312. Dyeing reaction: 20 min. Almost all cells (more than 90%) show a positive reaction.
  • E Untransfected HeLa cells (control for the specificity of the ⁇ -galactosidase reaction). Staining reaction: 3 h.
  • Example 16 Example 16
  • a 3.0 kb Sall fragment containing a single P. pyralis luciferase coding sequence under the control of the RSV promoter was ligated from plasmid p22RSVLuc ⁇ and into the single Sall site of the Cl-7al cosmid clone to form concatamers.
  • Cl-7al contains a 37 kb human genomic DNA Sau3A fragment (rapid digest), coding for no obvious genes, cloned into the BamHI site of the cosmid vector pW15 (Stratagene)).
  • the ligation reaction product was then packaged in vitro and an aliquot of the phage particles obtained was infected in E.coli MN544 and plated on LB amp plates.
  • the recombinants were screened by colony hybridization using the 3.0 kb Sall fragment (2_p_rnarked by randomized priming) as a hybridization probe and a number of positives were analyzed by restriction mapping.
  • a cosmid construct (CosLuc) containing a single copy of the Sall insert was grown and purified on a cesium gradient (total size: 48 kb).
  • a small control cosmid pWELuc (12 kb) was made by digesting CosLuc with Not I, religating, transforming bacteria and isolating the correct plasmid. This resulted in a 12 kb DNA molecule that lacked part of the human DNA insert and part of the CosLuc polylinker.
  • the plasmid pSPNeoLuc (8 kb) is the plasmid described in Example 5, which clones an RSV luciferase gene fragment (an Apal / Pvul fragment of pRSVL, cloned into the clal site of the pUC ⁇ locus).
  • HeLa cells (3 x 10 4 cells per 6 cm dish), covered with 1 ml DMEM + 2% FCS, were prepared with TfpL / DNA complexes, as described in the introduction to the examples, containing the stated amounts of hTfpL, free polylysine and DNA.
  • the incubation mixtures contained either 100 ⁇ M chloroquine (columns 1 and 2) or 10 ⁇ l adenovirus dl312, containing 5 ⁇ 1011 particles per ml (columns 3-12). After a 2 hour incubation at 37 ° C, 4 ml DMEM + 10% FCS were added to each dish. 24 hours later, the cells were harvested and luciferase activity measured. The results are shown in Fig. 22A.
  • the solution (2.5 ml) was flushed with argon and reacted with the exclusion of oxygen and argon with 42 ⁇ l of a solution of FITC-labeled polylysine (1 nmol) modified with 2.3 nmol mercaptopropionate groups (prepared as described in EP 388 758). After 18 h at room temperature, half of the solution was transferred to a centrifuge tube, carefully layered with 1 ml of a cesium chloride solution (density 1.33 mg / ml) and centrifuged for 2 h at 35000 rpm (SW60 rotor) at room temperature.
  • a cesium chloride solution density 1.33 mg / ml
  • the virus band was collected as a 200 ⁇ l cesium chloride fraction and diluted to 1 ml with HBS / 50% glycerol.
  • a DNA binding assay was carried out with 300 ⁇ l of the modified virus: the virus solution was diluted with 1 ml HBS and 100 ⁇ l solution of a 35s-labeled DNA (15 ng pRSVL, produced by nick translation) was added. As a control, the experiment was carried out in parallel with the same amount of unmodified virus dl312.
  • K562 cells (ATCC CCL 243) were suspended in RPMI 1640 medium (Gibco BRL plus 2 g sodium bicarbonate / 1) plus 10% FCS, 100 units / ml penicillin, 100 ⁇ l / ⁇ l streptomycin and 2 mM glutamine to a density of 500,000 cells / ml. 12 to 20 h before the transfection, the cells were placed in fresh medium containing 50 ⁇ M desferrioxamine (this measure was taken in order to achieve an increase in the transferrin receptor number).
  • the cells were collected, suspended in fresh medium containing 10% FCS plus 50 ⁇ M desferrioxamine (250,000 cells / ml) and placed in a 24-well dish in 2 ml portions.
  • Specified amounts of pCMVL-DNA (6, 0.6, 0.06 ⁇ g) in 100 ⁇ l HBS were mixed with 50 ⁇ l polylysine adenovirus (pLAdeno) or corresponding quantities (35 ⁇ l) control adenovirus dl312. After 20 min, appropriate amounts (12, 1.2, 0.12 ⁇ g) of TfpL190B conjugate in 150 ⁇ l HBS were added. After a further 20 minutes, the mixture was added to the K562 cells.
  • the cells were incubated at 37 ° C for 24 hours and then harvested for the luciferase assay. Luciferase activity was determined as indicated in the previous examples. The values given in Fig. 24 represent the total luciferase activity of the transfected cells.
  • One way to check the activity of a polylysine-virus conjugate is to check the conjugate for its ability to transport very small amounts of DNA (less than ⁇ g). Increased DNA transfer capacity was expected when the adenovirus was bound directly to the polylysine-condensed DNA because of the internalizing factors (Transferrin and adenovirus fiber protein) are directly associated with the DNA to be transported. In order to check the accuracy of this assumption, a constant amount of the polylysine-adenovirus conjugate (2.5 ⁇ l, approx. 5xl ⁇ 7 virus particles) was complexed with different amounts (3 ⁇ g to 0.0003 ⁇ g) of reporter plasmid in 475 ⁇ l HBS.
  • ATZBLATT coupled virus has its gene transfer capacity at both 0.003 and 0.0003 ⁇ g DNA. This amount of DNA corresponds to approx. 100 DNA molecules / cell and approx. 1 virus particle / DNA molecule.
  • the reaction mixture for the enzymatic coupling consists of 1150 ⁇ l of the virus elution fraction, 0.5 nmol guinea pig liver transglutaminase (TG) (Sigma), 2 nmol or 20 nmol polylysine290, 10 mM CaCl2 and reaction buffer in a final volume of 1500 ⁇ l.
  • the reaction was carried out at 37 ° C. for 1 hour and then stopped by adding 30 ⁇ l of 0.5M EDTA. To check the specificity of the coupling, reaction batches without transglutaminase were also carried out.
  • Unincorporated polylysine was separated from the viruses by centrifugation over a CsCl gradient (density 1.33 g / ml; 170,000 x g, 2 hours). The fraction containing the viruses was drawn off and mixed with the same volume of glycerol and frozen in liquid nitrogen and kept at -70 ° C.
  • REPLACEMENT3LATT The reaction was carried out with polylysine labeled with 125j using Bolton-Hunter reagent (Amersham) as described above. After the CsCl gradient centrifugation, the virus fraction was drawn off and separated using a further CsCl gradient. The gradient was then fractionated and the radioactivity in all fractions was determined using a scintillation counter. As shown in FIG. 26, it was found that in the reaction mixture with TG (dl312 / TG-pL) radioactive polylysine was enriched in the virus fraction (virus). In the control batch without TG (dl312 / pL) there was no accumulation of radioactive polylysine in the virus fraction.
  • ⁇ 10 5 cells (mouse hepatocytes; ATCC No.: TIB 73) were sown in DMEM with 10% heat-inactivated fetal calf serum (FCS), 2 mM glutamine, 100 IU / ml penicillin and 100 ⁇ g / ml streptomycin in 6 cm culture dishes.
  • FCS heat-inactivated fetal calf serum
  • Virus-DNA-Transferrin Complexes 50 ⁇ l of the respective polylysine-modified virus fraction were mixed with 6 ⁇ g of the DNA plasmid pCMVL in 10 ⁇ HBS and incubated for 20 min at room temperature. Then 8 ⁇ g of mouse transferrin-polylysine29OB (mTfpL) was added to the mixture and incubated for a further 10 min.
  • mTfpL mouse transferrin-polylysine29OB
  • the virus-DNA-transferrin complexes were mixed with 1.5 ml medium (DMEM with 2% FCS, 2 mM glutamine and antibiotics) and added to the cells after the old medium was removed. After incubation for 2 h at 37 ° C, 2 ml of DMEM with 10% FCS, glutamine and antibiotics were added to the cells. After a further cultivation phase of 2 hours, the entire medium was removed and 4 ml of fresh DMEM with 10% FCS, glutamine and antibiotics were added to the cells.
  • DMEM 2% FCS, 2 mM glutamine and antibiotics
  • the cells were harvested and the luciferase assay performed as described above.
  • complexes were also used for the transfection with the starting preparation of adenoviruses, which were neither treated with TG nor with polylysine (dl312). This preparation resulted in 4403000 light units.
  • Polylysine modified adenoviruses compared to unmodified adenoviruses, especially with low amounts of DNA
  • transfection was carried out as described in Example 3c), with 50 ⁇ l of the adenovirus fraction dl312 / TG-20 nmol pL and 6 ⁇ g pCMVL / 8 ⁇ g mTfpL, 0.6 ⁇ g pCMV-Luc / 0.8 ⁇ g mTfpL or 0.06 ⁇ g pCMVL / 0.08 ⁇ g for complex formation mTfpL were used.
  • transfections were also carried out with 6 ⁇ g, 0.6 ⁇ g, 0.06 ⁇ g pCMVL / mTfpL complexes and unmodified adenoviruses (dl312). It was found that the complexes with polylysine-modified adenoviruses still gave high expression values even with low amounts of DNA, while expression was greatly reduced in the case of unmodified adenoviruses (FIG. 28).
  • the biotinylation of the virus was possible in one qualitative detection after drops of various dilutions on cellulose nitrate membrane can be detected: after drying at 80 ° C / 2 h in a vacuum drying cabinet, blocking with BSA, incubation with streptavidin-conjugated alkaline phosphatase (BRL), washing and 1 h incubation with the developing solution NBT / X-phosphate (nitro blue tetrazolium salt / 5-bromo-4-chloro-3-indolyl phosphate, toluidine salt; Boehringer Mannheim) a positive color reaction was found.
  • NBT / X-phosphate nitro blue tetrazolium salt / 5-bromo-4-chloro-3-indolyl phosphate, toluidine salt; Boehringer Mannheim
  • the modified protein was reacted with 3-mercaptopropionate-modified polylysine (75 nmol, average chain length 290 lysine monomers, modified with 190 nmol mercaptopropionate linker) in 2.6 ml of 100 mM HEPES pH 7.9, 150 mM NaCl under an argon atmosphere. Conjugates were created using
  • HeLa cells were grown in 6 cm culture dishes as indicated in Example 1. The transfections were carried out at a density of 300,000 cells / plate. Before transfection, the cells were incubated with 1 ml of fresh medium containing 2% FCS.
  • 0.6 ⁇ g pCMVL-DNA in 67 ⁇ l HBS were mixed with 0.3 ⁇ g streptavidin-polylysine in 33 ⁇ l HBS.
  • 65 ⁇ l of biotinylated adenovirus or, as a control, corresponding amounts of adenovirus dl312 (30 ⁇ l, starting virus for the modification) were added.
  • the complex mixtures (“biotinAdV / complex A” or “control AdV "(see FIG. 29) were left to stand for a further 20 min and then diluted to 500 ⁇ l with HBS.
  • the alternative complex formation was carried out by first mixing 65 ⁇ l biotinylated adenovirus with 0.3 ⁇ g streptavidin-polylysine in 50 ⁇ l HBS after 20 min 0.6 ⁇ g of pCMVL-DNA in 50 ⁇ l of HBS were added The complex mixture (“biotinAdV / complex B”) was left to stand for a further 20 min and then diluted to 500 ⁇ l with HBS.
  • the mixtures were added to the cells.
  • the cells were incubated for 2 hours at 37 ° C., then 2.5 ml of fresh medium with the addition of 10% FCS were added.
  • the cells were incubated for 24 hours at 37 ° C. and then harvested for the luciferase assay.
  • the luciferase activity was determined as indicated in the previous examples, and the values shown in Figure 29 represent the total luciferase activity of the transfected cells.
  • K562 cells were suspended in RPMI 1640 medium (Gibco BRL, plus 2 g sodium bicarbonate per liter) + 10% FCS, 100 units / ml penicillin, 100 ⁇ g / ml streptomycin and 2 mM glutamine to a cell density of 500,000 cells / ml grown. 16 h before the transfection, the cells were placed in fresh medium containing 50 ⁇ M deferrioxamine (Sigma). The morning after the transfection, the cells were taken up to 250,000 cells / ml in fresh medium containing 10% FCS + 50 ⁇ M deferrioxamine and 2 ml per well were placed in a 24-well plate.
  • biotinylated adenovirus with 400 ng streptavidin-modified Incubate polylysine in 20 ul HBS for 20 min. Then 20 ul HBS containing 6 ug pCMVL were added. After an incubation period of 20 min, 7 ⁇ g mouse transferrin-polylysine conjugate (mTfpL) in 160 ⁇ l HBS were added and the entire mixture was incubated for a further 20 min.
  • mTfpL mouse transferrin-polylysine conjugate
  • the bone marrow cells were obtained from the culture medium by centrifugation at 100 x g for 8 minutes.
  • the cell precipitate was taken up in 3 ml of culture medium containing 2% FCS and 250 ⁇ l of the adenovirus-transferrin-polylysine / DNA complexes and grown in a new T25 bottle at 37 ° C. for 3 hours. Then 3 ml and after 2 h a further 6 ml of culture medium containing 10% FCS were added.
  • the cells were harvested and examined for luciferase expression as in the other examples.
  • the transfection resulted in luciferase activity corresponding to 310 x 103 light units / 100 ⁇ g total cell protein.
  • Cells from the cell line designated GI-ME-N were, as described in Example 16, transfected with the 48 kb cosmid with the stated amounts of hTfpL, free polylysine and DNA.
  • the cell line designated GI-ME-N was, as described in Example 16, transfected with the 48 kb cosmid with the stated amounts of hTfpL, free polylysine and DNA.
  • the cell line designated GI-ME-N were, as described in Example 16, transfected with the 48 kb cosmid with the stated amounts of hTfpL, free polylysine and DNA.
  • REPLACEMENT LEAF Incubation mixtures either 100 ⁇ M chloroquine (columns 3 and 4) or 10 ⁇ l adenovirus dl312, containing 5 x 10 - * - 1 particles per ml (columns 5 and 6).
  • the last two samples contained 15 ⁇ l biotinylated adenovirus dl312 (1 ⁇ 10 11 particles), incubated for 30 min with streptavidin-polylysine (0.8 ⁇ g, prepared as in Example 19) in 150 ⁇ l HBS.
  • 1 ° AE cells were obtained from patient nasal polyp samples as described by Yankaskas et al., 1987.
  • the tissues were in sterile saline washed, then transported to the laboratory in Joklik's Minimum Essential Medium (MEM) plus antibiotics (penicillin 50 U / ml, streptomycin 50 ⁇ g / ml, gentamicin 40 ⁇ g / ml) at 4 ° C.
  • Cartilage and excess submucosal tissue were exposed and the epithelial layers in protease solution (Sigma, type 14, 0.1 mg / dl) in MEM at 4 ° C for 16 to 48 h.
  • FBS fetal bovine serum
  • the cells were then treated with transferrin-polylsin conjugates (hTfpL) and a plasmid coding for luciferase (pRSVL) as a reporter gene.
  • hTfpL transferrin-polylsin conjugates
  • pRSVL plasmid coding for luciferase
  • biotinylated adenoviruses were used.
  • the cells treated with this conjugate showed a significantly stronger expression than background values (addition of conjugates 2585753 ⁇ 453585 light units).
  • Example 23 Gene transfer in hepatocytes and in blood cells
  • Transfection of Tissue Culture Cells Cells from the BNL CL.2 cell line were grown as described in Example 6. HeLa cells and hepatocytes were grown in 6 cm petri dishes. The transfection was carried out at a cell density of approx. 3 ⁇ 105 cells per dish. Before the transfection, the standard culture medium was replaced by 1 ml of fresh medium with 2% FCS.
  • Biotinylated adenoviruses (approx. 109 PFUs, see Example 19 a) and 19 b)) were reacted with 800 ng streptavidinilated polylysine in 50 ⁇ l HBS. After 30 min at room temperature, 6 ⁇ g pCMVL-DNA in 170 ⁇ l HBS were added, incubated for 30 min and then 3 ⁇ g pL300 in 200 ⁇ l HBS added and after a further 30 min the solution was used for the transfection experiments.
  • Biotinylated adenoviruses (approx. 109 PFUs) were reacted with 800 ng streptavidinilated polylysine in 50 ⁇ l HBS. After 30 min at room temperature, 6 ⁇ g pCMVL-DNA in 170 ⁇ l HBS were added, incubated for 30 min and then 10 ⁇ g TfpL190B in 200 ⁇ l HBS added and after a further 30 min the solution was used for the transfection experiments.
  • ⁇ -galactosidase assay BNL CL.2 cells were sown on cover slips and the cells were transfected with the reporter plasmid pCMV- ⁇ gal (Lim and Chae, 1989) for 24 h. The ⁇ -galactosidase test was carried out 48 hours later as indicated in Example 15. a) The connection between DNA condensates and
  • Adenovirus greatly enhances luciferase reporter gene expression
  • Fig. 32 The result of the transfer of DNA into hepatocytes using binary and ternary DNA complexes is shown in Fig. 32: The gene transfer effect was enhanced in the presence of free adenovirus.
  • Columns pLAdenoV / TfpL show the results of transfections with adenovirus conjugated to polylysine using transglutaminase and then allowed to react with DNA that neutralized some of the negative charges. Transferrin-polylysine was later added, which neutralized the remaining negative charges. In this way, a ternary complex adenovirus-polylysine / transferrin-polylysine / DNA was formed.
  • Cells of the human erythroleukemia cell line K562 contain approx. 150,000 transferrin receptors (Klausner et al., 1983 b). In the presence of chloroquine, these cells can also be extensively transfected with TfpL / reporter-DNA complexes in the absence of adenovirus (Fig. 33, TfpL), as previously reported by Cotten et al., 1990.
  • the same complexes provide relatively poor reporter gene expression (AdenoV / TfpL) in the presence of free adenovirus, but in the absence of chloroguin, presumably because K562 cells, like other blood cells (Silver et al., 1988; Horvath et al., 1988), deliver a low number Have adenovirus receptors. If the adenovirus is bound to polylysine via a biotin / streptavidin bridge and the reporter DNA is fully condensed by adding more polylysine to complete the binary complex (pLAdenoV / p), the transfection supported by adenovirus reaches medium values, presumably because the few adenovirus receptors are used efficiently.
  • the percentage is increased to approximately 0.2% (FIG. 34A).
  • free adenovirus about 5-10% of the cells express the reporter gene (Fig. 34B), while the ternary complexes with transglutaminase-modified virus lead to expression in most if not all cells (Fig. 34C).
  • the ternary complexes can be used at high dilution, the toxic effect observed at high doses of free (inactivated) adenovirus usually does not arise. It should be noted, however, that when the ternary complexes are used in high concentration to reach 100% of the tissue culture cells, a similar toxic effect becomes noticeable. The toxic effects can be due to residual viral gene activity or the endosomolytic properties of the added virus, or they are simply a consequence of the very high expression of the transfected gene.
  • a transfected reporter gene is transient, but persists for weeks in non-dividing hepatocytes
  • Ternary transport complexes pLAdenoV / TfpL
  • pLAdenoV / TfpL Ternary transport complexes
  • a 2/3 confluent hepatocyte cell culture was transfected with the luciferase reporter gene plasmid pCMVL as in the experiment shown in FIG. 34B, and the luciferase activity was measured at different times.
  • luciferase activity was highest after 3 days when the hepatocyte cell culture became confluent and the cells stopped dividing. Expression of the reporter gene persisted in the non-dividing cell culture without selection to maintain the gene and continued for at least 6 weeks, especially when psoralen-inactivated adenovirus was used to form the ternary complexes.
  • the chicken adenovirus CELO was tested for its suitability for enhancing DNA transfer in human HeLa cells analogous to the previous examples using the human adenovirus type 5.
  • the chicken adenovirus CELO (strain Phelps, serotype FAV-1.7, chicken kidney cell passage) was used.
  • the virus (2 ml) was passed through a PD-10 gel filtration column, equilibrated with 2.0 mM HEPES pH 7.3, 150 mM NaCl (HBS) + 10% glycerol, and 2 ml of the eluate were treated with 20 ⁇ l 1 mM NHS-LC-Biotin ( Pierce) reacted for 3 hours at room temperature.
  • the biotinylated virus was then dialyzed against 3 x 300 ml HBS + 40% glycerol at 4 ⁇ C and stored in aliquots at -70 ° C.
  • HeLa cells (5 x 105 cells per 6 cm dish) were incubated in 2 ml DMEM + 2% FCS with 6 ⁇ g of the plasmid pCMVL, complexed with polylysine (pLys) or transferrin-polylysine (TfpL) mixtures in 500 ⁇ l HBS (The complexes were preincubated for 30 min at room temperature). The samples were then added to the cells in the presence of the amounts of virus shown in FIG. 36.
  • pLys polylysine
  • TfpL transferrin-polylysine
  • the indicated amount of virus was preincubated with the indicated amount of streptavidin polylysine (StrpL) in 200 ⁇ l HBS for 30 min at room temperature before adding 6 ⁇ g of the plasmid pCMVL in 100 ⁇ l HBS were. After a 30 minute incubation at room temperature, the amount of material TfpL- at 37 C ⁇ specified was added to the cells. 2 hours later, 5 ml of DMEM + 10% FCS were added to the cells and the cells were harvested 24 hours later and prepared for the luciferase assay.
  • the CELO virus in free form increased the DNA transport in HeLa cells (columns 1-6).
  • the CELO virus was modified with biotin and contained in a complex with streptavidin, it was found that the virus, both with and without additional transferrin polylysine, enhanced DNA transfer to a comparable extent to that achieved with human adenovirus dl312.
  • the special line of HeLa cells shows a high binding capacity for polylysine / DNA complexes in the absence of transferrin (see
  • Luciferase activity of samples 1 and 4 in Fig. 36 It is therefore sufficient to include the CELO virus in a polylysine-DNA complex, the inclusion of the To do complex things.
  • C2C12 myoblasts (Blau et al., 1985; ATCC no .: CRL 1772) and G8 myoblasts (ATCC no .: CRL 1456) were in "high glucose DMEM” + 10% FCS myoblast cultures were subconfluent with approx. 5 x 10 5 Transfected cells per 6 cm dish.
  • Myotube cultures were prepared by plating myoblasts in 6 cm dishes (approx. 5 x 105 cells per dish) and exchanging the medium for "high glucose DMEM” + 2% horse serum when the cells reached confluence (Barr and Leiden, 1991; Dhawan et al., 1991). The myotubes were transfected 5 to 7 days later.
  • the transfection complexes were prepared as described in Example 19 using the indicated amounts of TfpL, StrpL and biotinylated adenovirus dl312.
  • the cells were harvested 20 h after the transfection and the luciferase activity was measured.
  • the luciferase activity shown in FIG. 37 relates to the entire cell sample. It was shown that both myoblast and myotube cultures could be transfected with high efficiency. After differentiation to myotubes, the transfection efficiency decreased less than 1 log (C2C12) or showed no significant decrease (G8).
  • the formation of myotubes occurred at a lower frequency in the G8 cell line, which is partly for that Lack of a detectable decrease in performance in the differentiated culture could be responsible.
  • C2C12 myotube cultures (like the myoblasts sown in 6 cm dishes, 5 x 10 ⁇ cells, differentiated in myotubes) were prepared as described in a).
  • pCMVß-gal DNA (6 ⁇ g) with 8 ⁇ g TfpL in the presence of 500 ⁇ l HBS
  • Coupled virus samples were prepared with pCMVLacz DNA (6 ⁇ g) complexed with 7 ⁇ g TfpL and 800 ng StrpL + 18 ⁇ l biotinylated adenovirus dl312 (1 ⁇ 1012 viruses per ml) in 500 ⁇ l HBS and the cells in 2 ml DMEM / 2% FCS added. After an incubation of 24 h, the cells were stained as described in Example 15 to determine the ⁇ -galactosidase activity.
  • the large skeletal muscles from the two hind legs of a 4-week-old male C57B1 / 6 mouse were isolated sterile in PBS and cut into approximately 5 mm pieces.
  • the tissue was suspended in 20 ml PBS, allowed to settle for approx. 2 min and the supernatant was aspirated. This washing was repeated 3 times.
  • the tissue was then mixed with 3.5 ml PBS plus 0.5 ml trypsin / EDTA, 0.5 ml 1% (w / v) collagenase, type 2 Sigma and 0.5 ml 1% BSA (fraction V in 4 mM CaCl2) and at 37 ° C for 30 min under frequent Allow gentle stirring to incubate.
  • the remaining tissue was allowed to settle, the supernatant was removed and mixed with 5 ml DMEM + 20% FCS.
  • the protease incubation was repeated 3-4 times until the tissue was completely dispersed.
  • the cell suspension was then passed through a cell sieve (Falcon) to remove aggregates and tissue fragments and centrifuged at 500 xg for 15 min.
  • the cell pellet was resuspended in 10 ml DMEM + 20% FCS and the fibroblasts removed by plating the cells on a 15 cm diameter uncovered tissue culture dish for 60 min.
  • the non-adherent cells were then carefully removed and plated on five laminin coated 10 cm tissue culture dishes with 15 ml DMEM + 20% FCS per dish.
  • the laminin-coated cell culture plates were prepared as follows: Cell culture dishes were coated with 0.025 mg / ml polylysine (MW 30,000-70,000, Sigma) in sterile water for 30 minutes at room temperature. The plates were rinsed 3 times with sterile water and air dried. The plates were then coated with 8 ⁇ g / ml laminin (EHS, Sigma) in water overnight at room temperature. The plates were then washed 3 times before sowing the cells.
  • the DNA complexes used for the transfection were prepared by diluting the stated amount of psoralen / UV-inactivated biotinylated adenovirus dl312 (prepared as in Example 19) in 150 ⁇ l HBS, adding 1 ⁇ g StrpL in 150 ⁇ l HBS and then incubating for 30 min at room temperature. HBS (100 ⁇ l) containing 6 ⁇ g pCMVL (referred to in the figure pCLuc) was then added to each sample and incubated for a further 30 min at room temperature.
  • Transfection into HeLa cells was performed using human adenovirus dl312 / StrpL / TfpL / DNA complexes that can enter the cells either through the adenovirus receptor or the transferrin receptor, or from CELO virus / StrpL / TfpL / DNA complexes that can enter via the transferrin receptor performed with comparable efficiency.
  • human adenovirus dl312 / StrpL / TfpL / DNA complexes that can enter the cells either through the adenovirus receptor or the transferrin receptor, or from CELO virus / StrpL / TfpL / DNA complexes that can enter via the transferrin receptor performed with comparable efficiency.
  • the transfer of DNA in C2C12 myoblasts with adenovirus dl312 complexes could be carried out efficiently, complexes with CELO viruses functioned poorly in these cells.
  • the transferrin receptor plays only a minor role in the transport of combination complexes into these
  • Biotinylated CELO virus was generated as previously described. Complexes containing 6 ⁇ g pCMVL plus the stated amounts of StrpL, TfpL, biotinylated wheat no agglutinin (WGA-B) and CELO virus were prepared as follows: Virus and WGA were diluted together in 150 ⁇ l HBS. StrpL
  • REPLACEMENT LEAF was also diluted in 150 ul HBS, the two solutions were mixed and incubated for 30 min at room temperature.
  • the DNA diluted in 100 ul HBS, was added to the StrpL / Virus / WGA solution, followed by another 30 minute incubation at room temperature. Finally, TfpL in 100 ⁇ l HBS was added to the mixture and the sample was incubated again at room temperature for 30 min.
  • the complexes were added to the C2C12 myoblasts (5 x 105 cells per 6 cm dish) in 2 ml DMEM + 2% FCS. One hour later, 5 ml DMEM + 10% FCS were added to the cells and 20 hours later the cells were prepared for the luciferase activity measurement.
  • the activity (light units) in each whole cell sample is shown in Fig. 40 (the DNA used in this example was pCMVL, designated pCLuc in the figure).
  • the myoblast and myotube cultures were prepared as described above. Transfections were carried out using 6 ⁇ g of a plasmid, containing a full length factor VIII cDNA (Wood et al., 1984; Eaton et al., 1986) and complexed according to the usual complex formation procedure with 5 or 15 ⁇ l biotinylated adenovirus (as indicated) plus 0.5 or 1 ⁇ g StrpL and 7 or 6 ⁇ g TfpL.
  • the DNA / virus complexes were applied to the cells in 2% FCS / DMEM. After a 4 hour incubation at 37 ° C, 3 ml of fresh DMEM + 10% FCS were added to each dish. 18 h later, the medium was harvested and examined using a COATEST (KABI, Pharmacia) test system with an international standard as a reference for the presence of factor VIII. The factor VIII amounts were applied as mUnits, formed in 24 h per 1 x 106 cells (FIG. 41).
  • Adenovirus wild type 300 was grown in HeLa cells, purified and, as described for adenovirus dl312, biotinylated. 1.2 ml of the virus was dialyzed against 3 x 300 ml of 5 mM MES (2- (N-morpholino) ethanesulfonic acid), 1 mM EDTA pH 6.25, 4 ° C. for 18 h. The material was then centrifuged at 27 K in a SW60 rotor for 30 min. The supernatant was carefully removed and the pellet resuspended in HBS / 40% glycerin.
  • HEPES, pH 7.4 and NaCl were added to the supernatant at 20 mM and 150 mM and both the pellet (containing the viral core proteins and the majority of the hexon capsid; in Fig. 42 "core”) and the supernatant fractions (containing the vertices) in FIG. 42 "vertices”) on their DNA transport activity in Movl3 mouse fibroblasts (Strauss and Jaenisch, 1992)
  • Examples 13 and 14 have shown that polylysine-conjugated peptides containing sequences derived from the N-terminus of the influenza virus hemagglutinin HA-2 subunit in DNA / transferrin-polylysine complexes significantly increase gene transfer by means of transferrin-polylysine .
  • the complexes were prepared as follows: biotinylated adenovirus dl312 (prepared as in Example 19; 2 ⁇ l, 6 ⁇ l or 18 ⁇ l; 10 --- 2 particles per ml) in 50 ⁇ l HBS were treated with streptavidin-polylysine (100 ng, 160 ng or 480 ng) mixed in 100 ul HBS. After an incubation of 30 min, a solution of 6 ⁇ g pCMVL in 200 ⁇ l HBS and after a further 30 min a solution of 3.8 ⁇ g (gal) 4pL (prepared as in Example 6) or 7 ⁇ g TfpL in 150 ⁇ l HBS was added.
  • the DNA complex solutions were added to 300,000 cells each (ATCC TIB73, ATCC TIB74, ATCC TIB75, ATCC TIB76), grown in 6 cm dishes in "high glucose DMEM" + 2% FCS.
  • the following cell culture steps and luciferase assays were performed as described in the previous examples, the gene expression values shown in Fig. 44 (after 24 h) were obtained.
  • Human-Ig and anti-human-Ig-polylysine conjugates were prepared as follows, the coupling being carried out analogously to methods known from the literature by introducing disulfide bridges after modification with succinimidyl-pyridyldithio-propionate (SPDP, Jung et al., 1981):
  • Poly (L) lysine 300 (average degree of polymerization of 300 lysine residues (Sigma)) was modified analogously with SPDP and brought into the form modified with free mercapto groups by treatment with dithiothreitol and subsequent gel filtration.
  • a solution of 12 nmoles of polylysine 300, modified with 29 nmoles of mercapto groups, in 0.3 ml of HBS was mixed with the above-mentioned modified anti-human-Ig in the absence of oxygen and left to stand at room temperature overnight.
  • the reaction mixture was brought to a content of 0.6 M NaCl by adding 5 M NaCl.
  • conjugates were isolated by ion exchange chromatography (Pharmacia, Mono S HR 5/5); After dialysis against 25 mM HEPES pH 7.3, corresponding conjugates consisting of 0.33 mg anti-human-Ig modified with 4 nmol of polylysine 300 were obtained (molar ratio 1: 2).
  • the conjugates were isolated by ion exchange chromatography (Mono S, Pharmacia, 50 mM HEPES pH 7.3, salt gradient 0.6 M to 3 M NaCl); After dialysis against HBS pH 7.3, corresponding conjugates consisting of 9 mg (57 nmol) antibody modified with 90 nmol polylysine 300 were obtained (molar ratio 1: 1.6).
  • the complexes were prepared as follows: biotinylated adenovirus dl312 (30 ul; 101 particles per ml) in 50 ul HBS were mixed with streptavidin-polylysine (800 ng) in 100 ul HBS; after 30 minutes of incubation, a solution of 9 ⁇ g pCMVL in 200 ⁇ l HBS was added. After a further 30 min, a solution of 5.1 ⁇ g polylysine (pLys450), 10.2 ⁇ g TfpL, 12 ⁇ g human-IgG-polylysine conjugate or 10 ⁇ g anti-human-Ig-polylysine conjugate in 150 ⁇ l HBS was added.
  • the DNA complexes were added to 10 ⁇ B lymphoblastoid cells (the cells were obtained from human peripheral blood mononuclear cells by immortalization with Epstein Barr virus, as described by Walls and Crawford, 1989, and in plates with 24 Wells grown in 1 ml RPMI 1640 + 2% FCS).
  • the further process of cell culture and the luciferase assays were carried out as described for the previous examples.
  • the gene expression values obtained (luciferase activity in light units) are shown in FIG. 45.
  • Rhinovirus HRV-2 was prepared and purified as described by Skern et al., 1984.
  • Photosensitive rhinovirus made by growing the virus in the presence of acridine orange was inactivated as described by Madshus et al., 1984.
  • Transferrin-polylysine / DNA complexes were prepared by adding a solution of 6 ⁇ g plasmid DNA pCMVL in 330 ⁇ l HBS (150 mM NaCl, 20 mM HEPES pH 7.3) with a solution of 8 ⁇ g TfpL290 was mixed in 170 ⁇ l HBS.
  • the DNA complexes were treated with 1.5 ml medium (DMEM + 2% FCS) and with 0.14 ⁇ g to 3.5 ⁇ g Rhinovirus HRV-2 (or inactivated HRV-2) mixed. The mixture was applied to NIH 3T3 cells (300,000 cells per 6 cm dish).
  • DNA combination complexes containing transferrin-polylysine and rhinovirus-polylysine conjugates were prepared as follows: a 100 ⁇ l solution of biotinylated rhinovirus HRV-2 (3.5 ⁇ g) in HBS was mixed with 1 ⁇ g StrpL in 100 ⁇ l HBS (the other virus concentrations were mixed with appropriate proportions).
  • the solution was mixed with 6 ⁇ g plasmid DNA in 150 ⁇ l HBS, incubated for a further 30 min at room temperature and then mixed with 6 ⁇ g TfpL290 in 150 ⁇ l HBS.
  • the DNA complexes were mixed with 1.5 ml of medium (DMEM + 2% FCS) and added to the NIH 3T3 cells (300,000 cells per 6 cm dish). The further treatment of the cultures and the luciferase assay were carried out as described in i) (FIG. 46B).
  • the DNA complexes were prepared by first mixing 30 ⁇ l adenovirus dl312 (approx. 109 PFUs) with 1 ⁇ g polylysine pLys450 (average chain length 450 monomers) in 170 ⁇ l HBS and then after 30 min at room temperature with 6 ⁇ g pCMVL -DNA in 170 ul HBS were added. After incubation for a further 30 min, the complexes were mixed with 9 ⁇ g TfpL190 in 170 ⁇ l HBS.
  • Complex formation B (control experiment): The DNA complexes were prepared by first 6 ⁇ g pCMVL-DNA in 170 ⁇ l HBS with 1 ⁇ g polylysine pLys450 in 170 ⁇ l HBS and, after 30 min at room temperature, then with 9 ⁇ g TfpL190 in 170 ⁇ l HBS was mixed. After incubation for a further 30 min, the complexes were mixed with 30 ⁇ l adenovirus dl312 (approx. 10 ⁇ PFUs).
  • the DNA complexes were prepared as described in Example 19. They contained 200 ⁇ l adenovirus dl312,
  • a male Sprague-Dawley rat (body weight 240 g) was anesthetized with avertine and a 4 cm long laparotomy was performed. The complexes were injected over a period of 2 min in the left lateral lobes of the liver. The laparotomy wound was then closed in layers. The rat was sacrificed 48 hours after injection of the complexes under ether anesthesia and the luciferase expression was measured in various liver samples.
  • luciferase activity of 5,615 light units / mg protein content of the liver homogenate in the area of the injection site; the total activity was 370,600 light units. No luciferase activity could be measured in the liver parts removed from the injection site.
  • the complexes were prepared as follows: 200 ⁇ l biotinylated adenovirus dl312, diluted with 200 ⁇ l HBS, were incubated with 6.4 ⁇ g streptavidin-modified polylysine in 400 ⁇ l HBS for 30 min at room temperature. Then 48 ⁇ g pCMVL in 800 ⁇ l HBS were added. After 30 min incubation, 48 ⁇ g TfpL in 900 ⁇ l HBS were added. To use the complexes, male Sprague-Dawley rats (250 g body weight) were anesthetized with avertine and the abdomen opened with a median incision.
  • the intestine was placed on the left side of the body and a 27 G needle connected to a tube and a 1 ml syringe was inserted into the bile duct.
  • the complexes were injected in 4 minutes.
  • the needle was withdrawn from the bile duct and the injection site with seal with a fibrin glue (Immuno).
  • the abdominal wound was closed with sutures and metal clips.
  • the rat was sacrificed and samples from various liver lobes were examined for luciferase expression.
  • the peak luciferase activity was 19,000 light units per mg protein; the calculated total expression in the whole liver was in the range of 2.7 x 10 6 light units.
  • the complexes were formed as described in Example 19. They consisted of 45 ⁇ l adenovirus dl312, 0.8 ⁇ g streptavidin-polylysine, 6 ⁇ g pCMVL and 24 ⁇ g mTfpL290 in a total volume of 150 ⁇ l HBS.
  • the complexes were injected into the tail vein of a male C3H / He mouse (age: 2 months) under anesthesia with avvertine. Immediately at the end of the injection, the tail vein was compressed at the proximal and distal tail ends so that the complex solution was retained in the injected tail vein section during the time of compression (25 min) and could not be flushed out with the blood.
  • the mouse 48 hours after the injection, the mouse was killed by cervical dislocation and the injected tail vein was prepared.
  • the expression of luciferase was measured in the homogenate of the tail vein section. This resulted in a luciferase activity of 2,600 light units / 3 cm tail vein.
  • Primary melanoma cells were isolated from a patient's surgically removed melanoma. For this purpose, the tumor was mechanically divided into RPMI 1640 cell culture medium with 5% FCS 2mM glutamine and antibiotic additive and the tissue fragments were pressed through a steel sieve. The tumor cells were then washed several times by centrifugation and resuspension and seeded in T25 cell culture bottles.
  • transfection with ternary complexes consisting of: 3 ⁇ l, 9 ⁇ l or 27 ⁇ l biotinylated adenovirus dl312 (1 x 10l2 / ml), 0.5 ⁇ g streptavidin-polylysine, 6 ⁇ g pCMVL and 7 ⁇ g TfpL290 (im Approach with 27 ⁇ l adenovirus: 1 ⁇ g streptavidin polylysine and 6 ⁇ g TfpL290) in a total volume of 500 ⁇ l HBS. 36 hours after the transfection, the cells were harvested and the luciferase activity (light units) was measured (see FIG. 47; the upper bar shows the results with the cells in suspension, the lower one with the adhering cells).
  • LDL-polylysine conjugates A solution of 10 mg (14.3 nmol) LDL (Low Density Lipoprotein, Sigma, L-2139, molecular weight 3,500,000, particle diameter approx. 26 nm) in 2 ml HBS was treated with 143 ⁇ l of a 10 mM added ethanolic solution of SPDP (1.43 ⁇ mol; Pharmacia) and reacted for 2 h at room temperature. Gel filtration on a Sephadex G25 column (14 x 140 mm) with HBS gave 3.2 ml of a solution of approx. 10 mg LDL, modified with 0.70 ⁇ mol pyridyldithiopropionate residues.
  • Poly (L) lysine with an average chain length of 300 monomers was modified with SPDP as described and brought into the form modified with mercapto groups by treatment with dithiothreitol and subsequent gel filtration.
  • the modified LDL solution described above was mixed with the solution of 0.33 ⁇ mol polylysine 300, modified with 0.76 ⁇ mol mercapto groups, in 4 ml 2 M NaCl, 0.5 M HEPES, pH 7.9, under argon and left to stand for 48 hours at room temperature.
  • the reaction solution was diluted to 32 ml with sterile water (NaCl concentration approx.
  • Human skin biopsies were placed in a 6 cm petri dish containing DMEM, 2 mM glutamine, 20% FCS and antibiotics. The biopsies were then chopped thoroughly with a surgical knife and grown in the presence of 3 ml of medium for 5 days. The cells were then washed with fresh medium (see above) and grown for a further 7 days. After this period, the cells were trypsinized and subcultured in new petri dishes. When the cells were almost confluent, they were trypsinized again and stored until transfection. For the transfection, the cells were thawed, sown in 6 cm petri dishes and in DMEM containing 2 mM Bred glutamine, 10% FCS and antibiotics.
  • the conjugates for the transfection were prepared as follows: 3 ⁇ l, 10 ⁇ l, 20 ⁇ l and 30 ⁇ l biotinylated adenovirus dl312 were incubated with 0.1 ⁇ g, 0.3 ⁇ g, 0.5 ⁇ g and 0.8 ⁇ g polylysine-modified streptavidin in 150 ⁇ l HBS for 30 min at room temperature . Then 6 ⁇ g of the plasmid pCMVß-gal in 170 ⁇ l HBS were added and the mixture was incubated for a further 30 min.
  • hTfpL Human transferrin-polylysine-DNA complexes
  • hTfpL Human transferrin-polylysine-DNA complexes
  • the rat Sigmodon hispidus (“Cotton Rat”) was used, which has been shown to be a suitable animal model for human adenoviral lung diseases (Pacini et al., 1984).
  • the animals were anesthetized with methoxyflurane. After a vertical cut in the ventral side of the neck, the trachea was bluntly dissected.
  • the complexes (250 to 300 ⁇ l; 3 ⁇ g plasmid DNA) were injected directly into the trachea of the animals inclined at an angle of 45 °. At the times indicated in FIG.
  • the animals were sacrificed by CO 2 and the trachea and the lungs were harvested en bloc after in situ rinsing with cold phosphate-buffered saline (PBS).
  • PBS cold phosphate-buffered saline
  • the lung tissue was homogenized in extraction buffer, the lysates standardized to total protein content and the luciferase gene expression measured as described.
  • the light units given refer to 1,250 ⁇ g total protein obtained from the lung lysates.
  • the experiments were carried out 3-4 times, the results are shown as mean values ⁇ SEM.
  • the peptides were on an automatic synthesizer (ABI 431A) using the solid phase method using p-alkoxybenyl alcohol resin (0.97 mmol / g) as the solid phase and Fmoc-protected amino acids.
  • the carboxy-terminal amino acid was bound to the resin via the symmetrical anhydride.
  • the following amino acids were coupled using the 1-hydroxybenzotriazole dicyclohexylcarbodiimide method.
  • the following side chain protecting groups were used: (Tr) Asn, (Trt) Cys [(-Bu) Cys in the case of EALA and GLF], (t-Bu) Glu, (Trt) His, (t-Bu) Ser.
  • the peptides had the following sequences:
  • EALA (SEQ ID NO: 5) Trp Glu Ala Ala Leu Ala Glu Ala Leu Ala Glu Ala Leu Ala Glu His Leu Ala Glu Ala Leu Ala Glu Ala Leu Glu Ala Leu Glu Ala Leu Ala Ala Gly Gly Ser Cys
  • GLF (SEQ ID NO: 6) Gly Leu Phe Gly Ala Leu Ala Glu Ala Leu Ala Glu Ala Leu Ala Glu His Leu Ala Glu Ala Leu Ala Glu Ala Leu Glu Ala Leu Glu Ala Leu Ala Ala Gly Gly Ser Cys
  • GLF-II (SEQ ID NO: 7) Gly Leu Phe Gly Ala Leu Ala Glu Ala Leu Ala Glu Ala Leu Ala Glu Ala Leu Ala Glu Ala Leu Glu Ala Leu Ala Ala Gly Gly Ser Cys GLF-delta (SEQ ID NO: 8) Gly Leu Phe Glu Leu Ala Glu Ala Leu Ala Glu Ala Leu Ala Glu Ala Leu Ala Glu Ala Leu Ala Glu Ala Leu Glu Ala Leu Ala Ala Gly Gly Ser Cys
  • EALA-Inf (SEQ ID NO: 9) Gly Leu Phe Gly Ala Ile Ala Gly Phe Ile Glu Asn Gly Trp Glu Gly Leu Ala Glu Ala Leu Ala Glu Ala Leu Glu Ala Leu Ala Ala Gly Gly Ser Cys
  • EALA-P50 (SEQ ID NO: 10) Gly Leu Phe Glu Ala Ile Glu Gly Phe Ile Glu Asn Gly Trp Glu Gly Leu Ala Glu Ala Leu Ala Glu Ala Leu Glu Ala Leu Ala Ala Gly Gly Ser Cys
  • the peptides were cleaved from the resin and the side chain protecting groups, with the exception of (t-Bu) Cys, were treated by treating 100 mg of the peptide-loaded solid phase with 3 ml of a mixture of phenol / ethanedithiol / thioanisole / water / trifluoroacetic acid 0.75: 0.25: 0.5 : 0.5: 10 removed for 1.5 h at room temperature. The crude peptides were precipitated in ether and washed twice.
  • the St-Bu protected peptides EALA and GLF were dissolved in a small volume of 1 M triethylammonium bicarbonate (TEAB) pH 8, diluted to 100 mM TEAB and further purified by reverse phase HPLC on a Nucleosil 500-5C4 column (0.1% TFA / acetonitrile Gradient), both peptides eluted at about 50% acetonitrile.
  • TEAB triethylammonium bicarbonate
  • the crude peptides (5 mg) were dissolved in 100 ⁇ l 100 mM TEAB, pH 8, containing 1 ⁇ l ⁇ -mercaptoethanol and by gel filtration (Sephadex G-25, 100 mM TEAB, 0.5 mM EDTA) and freeze-drying or ion exchange chromatography (Mono Q Pharmacia, 20 mM HEPES, pH 7.3, gradient 0 to 3 M NaCl; the peptide eluted at 1.5 M NaCl).
  • the homodimer of P50 (P50 dim) was prepared by allowing equimolar amounts of P50-Cys- (2-pyridylthio) and P50-Cys-SH to react in 20 mM HEPES, pH 7.3, for 3 days at room temperature were.
  • the reaction mixture was separated on a Mono Q column (HR-5/5 Pharmacia, 20 mM HEPES pH 7.3, gradient 0.09 M to 3 M NaCl; the P50 dimer eluted at 1.1 M NaCl).
  • the heterodimer GLF-SS-P50 was prepared analogously by reaction of the peptide P50 (free mercaptoform) with pyridylthiomodified peptide GLF.
  • the ability of the synthetic peptides to break liposomes was measured by the escape of fluorescent dye from liposomes loaded with a self-quenching concentration of calcein.
  • the liposomes were prepared using the REV method ("reverse phase evaporation") (Szoka and Papahadjopoulos, 1978) with an aqueous phase of 100 mM calcein, 375 mM Na +, 50 mM NaCl, pH 7.3 and through a 100 nm polycarbonate filter (MacDonald et al., 1991) extruded to obtain a uniform size distribution.
  • the liposomes were separated from unincorporated material by gel filtration on Sephadex G-25 with an iso-osmotic buffer (200 mM NaCl, 25 mM HEPES, pH 7.3).
  • an iso-osmotic buffer 200 mM NaCl, 25 mM HEPES, pH 7.3.
  • the stock solution (6 ⁇ l / ml) was diluted in 2 ⁇ assay buffer (400 mM NaCl, 40 mM Na citrate).
  • Fresh human erythrocytes were washed several times with HBS and resuspended in 2 x assay buffer of suitable pH (300 mM NaCl, 30 mM Na citrate) at a concentration of 6.6 x 107 / ml. An aliquot of 75 ⁇ l was added to 75 ⁇ l of a serial dilution of the peptide in water in a 96-well microtiter plate (cone type) and incubated for 1 hour at 37 ° C. with constant shaking.
  • suitable pH 300 mM NaCl, 30 mM Na citrate
  • the DNA complexes were prepared by first mixing 6 ⁇ g pCMVL-DNA in 150 ⁇ l HBS with 4 ⁇ g TfpL290 in 150 ⁇ l HBS and then after 30 min at room temperature with 4 to 20 ⁇ g poly (L) lysine290 in 100 ⁇ l HBS . After a further 30 min incubation at room temperature, 0.3 to 30 ⁇ g peptide in 100 ⁇ l HBS were added and the mixture was incubated for a further 30 min. The optimal amount of endosomolytic agents was determined in pre-titrations, whereby the performance of the gene transfer obtained was measured (see Table 3: gene transfer in BNL CL.2 cells).
  • Adherent cell lines (BNL CL.2 hepatocytes or NIH 3T3 cells) were grown 1 to 2 days before the transfection in 6 cm dishes (DMEM medium with 10% FCS; 300,000 cells / dish). The medium was removed and 1.5 ml DMEM + 2% FCS and 500 ⁇ l DNA complexes were added. Alternatively, 0.5 ml DMEM + 6% FCS and 1.5 ml DNA complexes were used. After 4 h of incubation, 2 ul DMEM (18% FCS) were added. (It was found that, alternatively, the medium can be replaced with 4 ml DMEM with 10% FCS.) The cell harvest and the luciferase assays were performed 24 hours after the transfection, as described. The values shown for the light units represent the
  • FIG. 52A The DNA complexes were prepared by first mixing 6 ⁇ g pCMVL-DNA in 250 ⁇ l HBS with 4 ⁇ g TfpL290 in 250 ⁇ l HBS and then after 30 min at room temperature with 20 ⁇ g poly (L) lysine 290 in 750 ⁇ l HBS were transferred. After a further 30 min incubation at room temperature, the stated amounts of peptides in 250 ⁇ l HBS were added. After an incubation of a further 30 min, the complexes were mixed with 0.5 ml DMEM + 6% FCS and 450,000 cells were added.
  • FIG. 52A The DNA complexes were prepared by first mixing 6 ⁇ g pCMVL-DNA in 250 ⁇ l HBS with 4 ⁇ g TfpL290 in 250 ⁇ l HBS and then after 30 min at room temperature with 20 ⁇ g poly (L) lysine 290 in 750 ⁇ l HBS were transferred. After a further 30 min incubation at room temperature, the stated amounts
  • the DNA complexes were prepared by first mixing 6 ⁇ g pCMVL-DNA in 500 ⁇ l HBS with 4 ⁇ g TfpL290 in 250 ⁇ l HBS and standing for 30 min at room temperature. The specified amounts of peptides in 250 ⁇ l HBS were added to a 500 ⁇ l solution of 20 ⁇ g poly (L) lysine290 in HBS and immediately added to the TfpL / DNA mixture. After a further 30 min incubation at room temperature, the complexes were mixed with 0.5 ml DMEM + 6% FCS and added to 450,000 cells. The cell harvest 24 h after the transfection and the luciferase assays were carried out as described.
  • the peptides P50 dim and EALA-P50 showed the highest activity, EALA and GLF had medium activity, while P50 monomers and melittin had low activity.
  • This peptide is derived from Staphylococcus aureus ⁇ -toxin (SEQ ID NO: 3) Met Ala Gin Asp Ile Ile Ser Thr Ile Gly Asp Leu Val Lys Trp Ile Ile Asp Thr Val Asn Lys Phe Thr Lys Lys, from which it is known that Has specificity for the rupture of membranes at acidic pH, derived by extending this peptide by 10 additional lysines.
  • the DNA complexes were prepared by first mixing 6 ⁇ g pCMVL-DNA in 170 ⁇ l HBS with 4 ⁇ g TfpL290 in 170 ⁇ l HBS and then after 30 min at room temperature with approximately 3 ⁇ g peptide in 170 ⁇ l HBS. After a further 30 min incubation at room temperature, the complexes were mixed with 1.5 ml DMEM + 2% FCS and 450,000 BNL CL.2 hepatocytes were added. After 2 h, 2 ml of DMEM + 20% FCS were added. The cell harvest 24 h after the transfection and the measurement of the luciferase activity were carried out as in the previous examples. The luciferase activity corresponding to the total extract was 481,000 light units.
  • Example 39 The luciferase activity corresponding to the total extract was 481,000 light units.
  • the DNA complexes were prepared by first mixing 6 ⁇ g pCMVL-DNA in 170 ⁇ l HBS with 4 ⁇ g TfpL290 in 170 ⁇ l HBS and then after 30 min at room temperature with approx. 3 ⁇ g peptide mell or 5 ⁇ g mel2 in 170 ⁇ l HBS has been. After incubation for a further 30 min, the complexes were mixed with 1.5 ml DMEM + 2% FCS and 450,000 BNL CL.2 cells, which were grown as in Example 38, were added. The cell harvest 24 h after the transfection and the luciferase test were carried out as described. The luciferase activity, based on the total extract, was 9,200 light units (in the case of mell) and 9,400 light units (in the case of mel2).
  • HeLa cells (5 x 105 cells per 6 cm dish) were transfected with the plasmid pAD-CMVl-IFN, which is for human Interferon alpha2c coded under the control of the CMV enhancer / promoter sequence (the plasmid is described in DE 40 21 917 A.
  • pAD-CMVl-IFN was obtained by cloning the HindIII-Xbal IFN- ⁇ 2c insert in pAD-CMV1) .
  • Samples of 6 ⁇ g DNA in 330 ⁇ l HBS were mixed with 8 ⁇ g TfpL in 330 ⁇ l HBS and left at room temperature for 30 minutes.
  • Samples 6-10 contained only 4 ⁇ g TfpL, and after the first 30 min incubation, samples 6 and 7 were each given an aliquot of P16pL (20 ⁇ g) in 160 ⁇ l HBS and samples 8, 9 and 10 an aliquot of pLys 290 (20 ⁇ g) added. After a further 30 min incubation, aliquots of 160 ⁇ l HBS containing 10 ⁇ l (sample 8) or 50 ⁇ l (samples 9 and 10) free P16 were added (for the synthesis of P16 and P16pL see example 13).
  • samples 2, 7 and 10 contained 100 ⁇ M chloroquine
  • samples 3 and 4 contained 5 and 15 ⁇ l Adenovirus dl312 (1 x 101 particles / ml)
  • sample 5 contained 15 ⁇ l of the same psoralen-inactivated virus.
  • samples 11, 12 and 13 were treated with aliquots of virus as in Examples 3, 4 and 5.
  • the medium was removed 48 h after the transfection and replaced with 2 ml of fresh DMEM + 10% FCS. This medium was harvested 72 h after the transfection and an ELISA test for the determination of IFN- ⁇ was carried out, as described in DE 40 21 917 A.
  • the IFN- ⁇ amounts (in ng / ml) are shown in Fig. 54.
  • TfpL functioned only weakly when transferring the IFN gene into these cells.
  • the presence of chloroquine produced a detectable signal (approx. 7 ng / ml, sample 2), while adenovirus dl312 stimulated DNA transfer in a dose-dependent manner (samples 3 and 4).
  • Treatment of these cells with comparable amounts of virus in the absence of IFN-DNA complexes gave no detectable IFN signal (samples 11 and 12).
  • Endothelial cells swine aorta + / - +++ + / ⁇

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biotechnology (AREA)
  • Veterinary Medicine (AREA)
  • Wood Science & Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Virology (AREA)
  • Toxicology (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Cell Biology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
PCT/EP1992/002234 1991-09-30 1992-09-28 Zusammensetzung für das einbringen von nukleinsäure-komplexen in höhere eukaryotische zellen WO1993007283A1 (de)

Priority Applications (17)

Application Number Priority Date Filing Date Title
KR1019940700967A KR100241685B1 (ko) 1991-09-30 1992-09-28 핵산 복합체를 고등 진핵세포내로 도입시키기 위한 조성물
AU26526/92A AU671084B2 (en) 1991-09-30 1992-09-28 Composition for inserting nucleic acid complexes into higher eucaryotic cells
EP92920580A EP0607206B1 (de) 1991-09-30 1992-09-28 Zusammensetzung für das einbringen von nukleinsäure-komplexen in höhere eukaryotische zellen
CA002118816A CA2118816C (en) 1991-09-30 1992-09-28 Composition for introducing nucleic acid complexes into higher eucaryotic cells
AT92920580T ATE215990T1 (de) 1991-09-30 1992-09-28 Zusammensetzung für das einbringen von nukleinsäure-komplexen in höhere eukaryotische zellen
SK368-94A SK281682B6 (sk) 1991-09-30 1992-09-28 Kompozícia na transfekciu buniek, komplex ako súčasť kompozície, konjugát ako súčasť komplexu, endozomolytický peptid, spôsob tvorby konjugátu, spôsob vnášania nukleových kyselín do buniek, farmaceutický prípravok s jeho obsahom a transfekčná súprava
BR9206559A BR9206559A (pt) 1991-09-30 1992-09-28 Composição para introdução de complexos de ácido nucléico em células eucarióticas superiores
JP5506590A JPH10506001A (ja) 1991-09-30 1992-09-28 核酸複合体を高等真核生物の細胞に導入するための組成物
ES92920580T ES2173083T3 (es) 1991-09-30 1992-09-28 Composicion para la incorporacion de complejos de acidos nucleicos en celulas eucarioticas superiores.
PL92303106A PL180304B1 (pl) 1991-09-30 1992-09-28 Kompozycja do transfekowania in vitro komórek wyzszych eukariontówkompleksem kwasu nukleinowego PL PL PL PL PL PL
DE59209951T DE59209951D1 (de) 1991-09-30 1992-09-28 Zusammensetzung für das einbringen von nukleinsäure-komplexen in höhere eukaryotische zellen
DK92920580T DK0607206T3 (da) 1991-09-30 1992-09-28 Præparat til indføring af nucleinsyrekomplekser i højere eukaryote celler
RO94-00499A RO117861B1 (ro) 1991-09-30 1992-09-28 Complex de acid nucleic si compozitie pentru introducerea acestuia in celulele eucariote superioare
NO19941154A NO316744B1 (no) 1991-09-30 1994-03-29 Sammensetning for innforing av nukleinsyrekomplekser i hoyere eukaryotiske celler, kompleks, endosomolytisk peptid, fremgangsmater og transfeksjonssett
FI941474A FI941474A0 (fi) 1991-09-30 1994-03-30 Seos nukleiinihappo-kompleksien viemiseksi korkeampiin eukaryoottisiin soluihin
BG98718A BG62740B1 (bg) 1991-09-30 1994-04-14 Състав за въвеждане на комплекси на нуклеинови киселини в по-висши еукариотни клетки
HK98113354A HK1013104A1 (en) 1991-09-30 1998-12-14 Composition for inserting nucleic acid complexes into higher eucaryotic cells

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
US76778891A 1991-09-30 1991-09-30
US76803991A 1991-09-30 1991-09-30
US82710292A 1992-01-30 1992-01-30
US82710392A 1992-01-30 1992-01-30
US86475992A 1992-04-07 1992-04-07
US93778892A 1992-09-02 1992-09-02
US864,759 1992-09-02
US768,039 1992-09-02
US767,788 1992-09-02
US827,103 1992-09-02
US827,102 1992-09-02
US937,788 1992-09-02

Publications (1)

Publication Number Publication Date
WO1993007283A1 true WO1993007283A1 (de) 1993-04-15

Family

ID=27560281

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP1992/002234 WO1993007283A1 (de) 1991-09-30 1992-09-28 Zusammensetzung für das einbringen von nukleinsäure-komplexen in höhere eukaryotische zellen

Country Status (28)

Country Link
US (4) US5547932A (sl)
EP (2) EP0545016A1 (sl)
JP (1) JPH10506001A (sl)
CN (1) CN1059705C (sl)
AT (1) ATE215990T1 (sl)
AU (1) AU671084B2 (sl)
BG (1) BG62740B1 (sl)
BR (1) BR9206559A (sl)
CA (1) CA2118816C (sl)
CZ (1) CZ293141B6 (sl)
DE (1) DE59209951D1 (sl)
DK (1) DK0607206T3 (sl)
EE (1) EE03195B1 (sl)
ES (1) ES2173083T3 (sl)
FI (1) FI941474A0 (sl)
HK (1) HK1013104A1 (sl)
HU (2) HU218846B (sl)
IL (1) IL103171A (sl)
MX (1) MX9205543A (sl)
NO (1) NO316744B1 (sl)
NZ (1) NZ244306A (sl)
PL (1) PL180304B1 (sl)
PT (1) PT607206E (sl)
RO (1) RO117861B1 (sl)
SG (1) SG44680A1 (sl)
SI (1) SI9200236B (sl)
SK (1) SK281682B6 (sl)
WO (1) WO1993007283A1 (sl)

Cited By (82)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994006923A1 (en) * 1992-09-24 1994-03-31 The University Of Connecticut Modification of a virus to redirect infectivity and enhance targeted delivery of polynucleotides to cells
WO1994024299A1 (de) * 1993-04-08 1994-10-27 Boehringer Ingelheim International Gmbh Adenovirus für den transport von fremd-dna in höhere eukariotische zellen
EP0654268A1 (en) * 1993-10-19 1995-05-24 BOEHRINGER INGELHEIM INTERNATIONAL GmbH A method to reverse the phenotype of transformed cells by the transcription factor IRF-1
WO1995026411A2 (en) * 1994-03-25 1995-10-05 The Uab Research Foundation Composition and methods for creating syngeneic recombinant virus-producing cells
WO1995033062A2 (de) * 1994-05-30 1995-12-07 Boehringer Ingelheim International Gmbh Verfahren zum einbringen von fremdmaterial in höhere eukaryotische zellen
WO1996003517A1 (de) * 1994-07-26 1996-02-08 Boehringer Ingelheim International Gmbh Verfahren zur herstellung von viren und viralen vektoren
WO1996029423A1 (en) * 1995-03-20 1996-09-26 Baylor College Of Medicine Compositions and methods for inducing infection by retroviral vectors outside of their host range
DE19510344C1 (de) * 1995-03-22 1996-11-07 Boehringer Ingelheim Int Verwendung einer Tumorvakzine
WO1996040958A1 (en) * 1995-06-07 1996-12-19 Baylor College Of Medicine Nucleic acid transporters for delivery of nucleic acids into a cell
WO1997000965A2 (en) * 1995-06-23 1997-01-09 University Of Pittsburgh A lipidic vector for nucleic acid delivery
WO1997010851A2 (en) * 1995-09-07 1997-03-27 Opperbas Holding B.V. Nucleic acid delivery vehicle
EP0789708A1 (en) * 1994-09-28 1997-08-20 Apollon, Inc. Multifunctional molecular complexes for gene transfer to cells
WO1997030170A1 (de) * 1996-02-15 1997-08-21 Boehringer Ingelheim International Gmbh Zusammensetzung für die transfektion höherer eukaryotischer zellen
WO1997033998A1 (en) * 1996-03-14 1997-09-18 The Immune Response Corporation Targeted delivery of genes encoding interferon
US5712136A (en) * 1994-09-08 1998-01-27 Genvec, Inc. Adenoviral-mediated cell targeting commanded by the adenovirus penton base protein
DE19632532A1 (de) * 1996-08-13 1998-02-19 Boehringer Ingelheim Int Verfahren zur Herstellung von Säugetieren mit definierten genetischen Eigenschaften
US5736392A (en) * 1995-06-07 1998-04-07 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US5770442A (en) * 1995-02-21 1998-06-23 Cornell Research Foundation, Inc. Chimeric adenoviral fiber protein and methods of using same
US5846782A (en) * 1995-11-28 1998-12-08 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
EP0900849A1 (en) * 1996-10-31 1999-03-10 Shanghai Cancer Institute Receptor-mediated gene transfer system for targeting tumor gene therapy
WO1999029349A1 (en) * 1997-12-04 1999-06-17 Boehringer Ingelheim International Gmbh Transfection particles
US5965404A (en) * 1994-05-31 1999-10-12 Boehringer Ingelheim International Fmbh Method for introducing nucleic acids into higher eukaryotic cells
US6010871A (en) * 1994-09-29 2000-01-04 Ajinomoto Co., Inc. Modification of peptide and protein
US6020202A (en) * 1993-07-12 2000-02-01 Life Technologies, Inc. Composition and methods for transfecting eukaryotic cells
WO2000011019A1 (en) * 1998-08-21 2000-03-02 Felix Frey Conjugates of dna interacting groups with steroid hormones for use as nucleic acid transfection agent
US6051429A (en) * 1995-06-07 2000-04-18 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US6127525A (en) * 1995-02-21 2000-10-03 Cornell Research Foundation, Inc. Chimeric adenoviral coat protein and methods of using same
US6153596A (en) * 1996-12-18 2000-11-28 Emory University Polycationic oligomers
US6335016B1 (en) 1996-04-20 2002-01-01 Boehringer Ingelheim International Gmbh Chicken embryo lethal orphan (CELO) virus
US6455314B1 (en) 1998-09-11 2002-09-24 Genvec, Inc. Alternatively targeted adenovirus
US6468981B1 (en) 1994-07-29 2002-10-22 Emory University Compositions and methods for targeting pharmaceutically active materials to cells containing androgen receptors
US6599737B1 (en) 1998-04-30 2003-07-29 Cornell Research Foundation, Inc. Adenoviral vectors with tandem fiber proteins
WO2003015825A3 (en) * 2001-08-16 2003-07-31 Photobiotics Ltd Conjugate
US6914131B1 (en) 1998-10-09 2005-07-05 Chiron S.R.L. Neisserial antigens
US6913922B1 (en) 1999-05-18 2005-07-05 Crucell Holland B.V. Serotype of adenovirus and uses thereof
US6929946B1 (en) 1998-11-20 2005-08-16 Crucell Holland B.V. Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
US6951755B2 (en) 1994-09-08 2005-10-04 Genvec, Inc. Vectors and methods for gene transfer
WO2005093064A1 (ja) 2004-03-29 2005-10-06 Galpharma Co., Ltd. 新規ガレクチン9改変体タンパク質及びその用途
US6974695B2 (en) 2000-11-15 2005-12-13 Crucell Holland B.V. Complementing cell lines
WO2006042017A2 (en) 2004-10-06 2006-04-20 University Of Rochester Treatment of pulmonary hypertension using an agent that inhibits a tissue factor pathway
US7105346B2 (en) 1995-06-15 2006-09-12 Crucell Holland B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
DE102005023993A1 (de) * 2005-05-20 2006-11-23 TransMIT Gesellschaft für Technologietransfer mbH Nicht virales Vektorsystem zum Transport von Nukleinsäure in die Lunge
US7235233B2 (en) 2000-09-26 2007-06-26 Crucell Holland B.V. Serotype 5 adenoviral vectors with chimeric fibers for gene delivery in skeletal muscle cells or myoblasts
WO2008020335A2 (en) 2006-06-09 2008-02-21 Novartis Ag Immunogenic compositions for streptococcus agalactiae
EP1935979A2 (en) 1999-07-14 2008-06-25 Novartis Vaccines and Diagnostics S.r.l. Antigenic meningococcal peptides
EP1950297A2 (en) 2000-05-31 2008-07-30 Novartis Vaccines and Diagnostics, Inc. Compositions and methods for treating neoplastic disease using chemotherapy and radiation sensitizers
EP1953229A2 (en) 1998-10-15 2008-08-06 Novartis Vaccines and Diagnostics, Inc. Metastatic breast and colon cancer regulated genes
EP1961813A2 (en) 1998-12-16 2008-08-27 Novartis Vaccines and Diagnostics, Inc. Human cyclin-dependent kinase (hPNQALRE)
US7468181B2 (en) 2002-04-25 2008-12-23 Crucell Holland B.V. Means and methods for the production of adenovirus vectors
EP2039768A1 (en) 1996-11-13 2009-03-25 Novartis Vaccines and Diagnostics, Inc. Mutant forms of Fas ligand and uses thereof
EP2110667A1 (en) 2005-01-20 2009-10-21 University Of Rochester Thoredoxin interacting protein (TXNIP) as regulator of vascular function
US7749493B2 (en) 1998-07-08 2010-07-06 Crucell Holland B.V. Chimeric adenoviruses
EP2210945A2 (en) 1998-01-14 2010-07-28 Novartis Vaccines and Diagnostics S.r.l. Neisseria meningitidis antigens
WO2010107991A2 (en) 2009-03-18 2010-09-23 Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services Use of ixolaris, a tissue factor inhibitor, for the treatment and prevention of cancer
EP2251424A1 (en) 1999-05-19 2010-11-17 Novartis Vaccines and Diagnostics S.r.l. Antigenic neisserial peptides
EP2261342A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2267005A1 (en) 2003-04-09 2010-12-29 Novartis Vaccines and Diagnostics S.r.l. ADP-ribosylating toxin from Listeria monocytogenes
EP2270177A1 (en) 2001-03-27 2011-01-05 Novartis Vaccines and Diagnostics S.r.l. Streptococcus pneumoniae proteins and nucleic acids
EP2275551A2 (en) 1999-10-29 2011-01-19 Novartis Vaccines and Diagnostics S.r.l. Neisserial antigenic peptides
EP2275129A2 (en) 2000-01-17 2011-01-19 Novartis Vaccines and Diagnostics S.r.l. Outer membrane vesicle (OMV) vaccine comprising N. meningitidis serogroup B outer membrane proteins
EP2277894A1 (en) 2000-10-27 2011-01-26 Novartis Vaccines and Diagnostics S.r.l. Nucleic acids and proteins from streptococcus groups A & B
EP2278007A1 (en) 1999-04-30 2011-01-26 Novartis Vaccines and Diagnostics S.r.l. Conserved neisserial antigens
EP2278006A2 (en) 1997-11-06 2011-01-26 Novartis Vaccines and Diagnostics S.r.l. Neisserial antigens
EP2298796A2 (en) 2001-03-27 2011-03-23 Novartis Vaccines and Diagnostics S.r.l. Staphylococcus aureus proteins and nucleic acids
EP2335724A1 (en) 2001-12-12 2011-06-22 Novartis Vaccines and Diagnostics S.r.l. Immunisation against chlamydia trachomatis
WO2012092341A1 (en) 2010-12-28 2012-07-05 University Of Rochester Methods of modifying insulin signaling using biliverdin reductase (bvr) and bvr derived peptides
WO2013036875A1 (en) 2011-09-07 2013-03-14 Mount Sinai School Of Medicine Ceramidase and cell differentiation
US8673859B2 (en) 2007-03-20 2014-03-18 New York University GM-CSF cosmeceutical compositions and methods of use thereof
WO2014063155A1 (en) 2012-10-21 2014-04-24 University Of Rochester Thy1 (cd90) as a novel therapy to control adipose tissue accumulation
WO2014176259A1 (en) 2013-04-22 2014-10-30 Icahn School Of Medicine At Mount Sinai Mutations in pdgfrb and notch3 as causes of autosomal dominant infantile myofibromatosis
US8945884B2 (en) 2000-12-11 2015-02-03 Life Technologies Corporation Methods and compositions for synthesis of nucleic acid molecules using multiplerecognition sites
US8951768B2 (en) 2005-05-04 2015-02-10 Kineta Two, Llc Mutations in OAS1 genes
US9090947B2 (en) 2003-10-23 2015-07-28 Kineta Two, Llc Detection of mutations in a gene associated with resistance to viral infection, OAS1
US9267163B2 (en) 2000-02-28 2016-02-23 Glaxosmithkline Biologicals Sa Hybrid expression of neisserial proteins
US9358300B2 (en) 1998-11-12 2016-06-07 Life Technologies Corporation Transfection reagents
US9534252B2 (en) 2003-12-01 2017-01-03 Life Technologies Corporation Nucleic acid molecules containing recombination sites and methods of using the same
WO2017079442A1 (en) 2015-11-04 2017-05-11 Icahn School Of Medicine At Mount Sinai Methods of treating tumors and cancer, and identifying candidate subjects for such treatment
WO2017189730A1 (en) 2016-04-26 2017-11-02 Icahn School Of Medicine At Mount Sinai Treatment of hippo pathway mutant tumors and methods of identifying subjects as candidates for treatment
EP3329932A1 (en) 2009-06-10 2018-06-06 New York University Immunological targeting of pathological tau proteins
US10195280B2 (en) 2014-07-15 2019-02-05 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
EP3553168A1 (en) 2010-11-12 2019-10-16 Georgetown University Immortalization of epithelial cells and methods of use
US10543232B2 (en) 2014-05-14 2020-01-28 Targimmune Therapeutics Ag Polyplex of double-stranded RNA and polymeric conjugate

Families Citing this family (334)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5354844A (en) * 1989-03-16 1994-10-11 Boehringer Ingelheim International Gmbh Protein-polycation conjugates
US5981273A (en) * 1991-09-30 1999-11-09 Boehringer Ingelheim Int'l. Gmbh Composition comprising an endosomolytic agent for introducing nucleic acid complexes into higher eucaryotic cells
NZ244306A (en) * 1991-09-30 1995-07-26 Boehringer Ingelheim Int Composition for introducing nucleic acid complexes into eucaryotic cells, complex containing nucleic acid and endosomolytic agent, peptide with endosomolytic domain and nucleic acid binding domain and preparation
CA2131620A1 (en) * 1992-03-20 1993-09-30 Louis C. Smith A dna transporter system and method of use
ES2221920T3 (es) * 1992-04-03 2005-01-16 The Regents Of The University Of California Sistema de liberacion de polinucleotidos de autoensamblaje que comprende un peptido cationico anfipatico.
HUT73383A (en) * 1993-03-19 1996-07-29 Boehringer Sohn Ingelheim Process for preparing cancer vaccines
DE4335025A1 (de) * 1993-10-14 1995-04-20 Boehringer Ingelheim Int Endosomolytisch wirksame Partikel
US5928944A (en) * 1994-02-04 1999-07-27 The United States Of America As Represented By The Department Of Health And Human Services Method of adenoviral-medicated cell transfection
MX9603866A (es) * 1994-03-18 1997-03-29 Boehringer Ingelheim Int Procedimiento para el tratamiento de celulas eucarioticas.
US5670347A (en) 1994-05-11 1997-09-23 Amba Biosciences Llc Peptide-mediated gene transfer
US5525606A (en) 1994-08-01 1996-06-11 The United States Of America As Represented By The Department Of Health And Human Services Substituted 06-benzylguanines and 6(4)-benzyloxypyrimidines
US5965541A (en) * 1995-11-28 1999-10-12 Genvec, Inc. Vectors and methods for gene transfer to cells
US5962311A (en) * 1994-09-08 1999-10-05 Genvec, Inc. Short-shafted adenoviral fiber and its use
US6221959B1 (en) 1994-11-18 2001-04-24 Supratek Pharma, Inc. Polynucleotide compositions
US6359054B1 (en) 1994-11-18 2002-03-19 Supratek Pharma Inc. Polynucleotide compositions for intramuscular administration
US6008202A (en) * 1995-01-23 1999-12-28 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5795587A (en) * 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
AU5103996A (en) * 1995-03-09 1996-10-02 Bavarian Nordic Research Institute A/S Vectors carrying therapeutic genes encoding antimicrobial peptides for gene therapy
US6420549B1 (en) 1995-06-06 2002-07-16 Isis Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
US20030069173A1 (en) * 1998-03-16 2003-04-10 Life Technologies, Inc. Peptide-enhanced transfections
ATE278794T1 (de) * 1995-06-15 2004-10-15 Crucell Holland Bv Verpackungssysteme für humane, menschliche adenoviren, zur verwendung in die gentherapie
GB9515356D0 (en) * 1995-07-26 1995-09-20 Medical Res Council Improvements in or relating to delivery of nucleic acid
US5770720A (en) * 1995-08-30 1998-06-23 Barnes-Jewish Hospital Ubiquitin conjugating enzymes having transcriptional repressor activity
CA2190304A1 (en) * 1995-12-15 1997-06-16 Elazar Rabbani Property effecting and/or property exhibiting compositions for therapeutic and diagnostic uses
AU2112697A (en) * 1996-02-09 1997-08-28 Pi-Wan Cheng Receptor ligand-facilitated delivery of biologically active molecules
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
DE69725878T2 (de) 1996-08-13 2004-07-29 Chiron Corp. (N.D.Ges.D. Staates Delaware), Emeryville Zusammensetzungen zur polynukleotidabgabe
US5948681A (en) * 1996-08-14 1999-09-07 Children's Hospital Of Philadelphia Non-viral vehicles for use in gene transfer
US5965441A (en) * 1996-11-13 1999-10-12 The General Hospital Coporation HSV/AAV hybrid amplicon vectors
US20060002949A1 (en) 1996-11-14 2006-01-05 Army Govt. Of The Usa, As Rep. By Secretary Of The Office Of The Command Judge Advocate, Hq Usamrmc. Transcutaneous immunization without heterologous adjuvant
US5980898A (en) 1996-11-14 1999-11-09 The United States Of America As Represented By The U.S. Army Medical Research & Material Command Adjuvant for transcutaneous immunization
US6797276B1 (en) 1996-11-14 2004-09-28 The United States Of America As Represented By The Secretary Of The Army Use of penetration enhancers and barrier disruption agents to enhance the transcutaneous immune response
JP4289687B2 (ja) * 1997-03-14 2009-07-01 ザ チルドレンズ ホスピタル オブ フィラデルフィア 血友病の治療のための遺伝子治療で使用する方法と組成物
US20040009166A1 (en) * 1997-04-30 2004-01-15 Filpula David R. Single chain antigen-binding polypeptides for polymer conjugation
CA2288992C (en) * 1997-04-30 2012-06-12 Enzon, Inc. Single-chain antigen-binding proteins capable of glycosylation, production and uses thereof
US6635623B1 (en) 1997-06-13 2003-10-21 Baylor College Of Medicine Lipoproteins as nucleic acid vectors
US20050096288A1 (en) * 1997-06-13 2005-05-05 Aragene, Inc. Lipoproteins as nucleic acid vectors
DE19726186A1 (de) * 1997-06-20 1998-12-24 Boehringer Ingelheim Int Komplexe für den Transport von Nukleinsäure in höhere eukaryotische Zellen
US6172211B1 (en) 1997-07-11 2001-01-09 Boehringer Ingelheim International Gmbh Nucleic acid encoding tag7 polypeptide
EP1007099A4 (en) * 1997-07-11 2004-11-24 Univ Brandeis METHOD FOR INDUCING APOPTOSE BY LOWERING THE THIAMINE MIRROR
US7923250B2 (en) 1997-07-30 2011-04-12 Warsaw Orthopedic, Inc. Methods of expressing LIM mineralization protein in non-osseous cells
DE69840361D1 (de) 1997-07-30 2009-01-29 Univ Emory Neue knochenmineralisierungsproteine, dna, vektoren, expressionssysteme
WO1999007723A1 (en) * 1997-08-07 1999-02-18 University Of Maryland, Baltimore Nucleic acid uptake and release vehicle
US20030125278A1 (en) * 1997-08-13 2003-07-03 Tang De-Chu C. Immunization of animals by topical applications of a salmonella-based vector
US6706693B1 (en) 1997-08-13 2004-03-16 The Uab Research Foundation Vaccination by topical application of genetic vectors
US6197332B1 (en) 1997-08-13 2001-03-06 Chiron Corporation Lipid-conjugated polyamide compounds and related compositions and methods thereof
US20030045492A1 (en) * 1997-08-13 2003-03-06 Tang De-Chu C. Vaccination by topical application of recombinant vectors
US6716823B1 (en) 1997-08-13 2004-04-06 The Uab Research Foundation Noninvasive genetic immunization, expression products therefrom, and uses thereof
US6348450B1 (en) 1997-08-13 2002-02-19 The Uab Research Foundation Noninvasive genetic immunization, expression products therefrom and uses thereof
US5998386A (en) 1997-09-19 1999-12-07 Feldman; Arthur M. Pharmaceutical compositions and method of using same for the treatment of failing myocardial tissue
WO1999019500A1 (en) * 1997-10-10 1999-04-22 The Government Of The United States Of America, Represented By The Secretary, Department Of Healt H And Human Services Complex of biotinylated viral vector and ligand for targeted gene delivery
US6734338B1 (en) 1997-11-14 2004-05-11 Cedars-Sinai Medical Center Transfection, storage and transfer of male germ cells for generation of transgenic species and genetic therapies
US7294755B1 (en) 1997-11-14 2007-11-13 Cedars-Sinai Medical Center Genetic modification of male germ cells for generation of transgenic species and genetic therapies
JP2001523468A (ja) * 1997-11-14 2001-11-27 セダーシナイ メディカル センター トランスジェニック種の産生のための雄性生殖細胞のトランスフェクションおよび移入
CA2317549C (en) * 1998-01-05 2006-04-11 University Of Washington Composition for enhancing transport through lipid-containing membranes, and uses thereof
US7244714B1 (en) * 1998-06-12 2007-07-17 Aradigm Corporation Methods of delivering aerosolized polynucleotides to the respiratory tract
US6509323B1 (en) 1998-07-01 2003-01-21 California Institute Of Technology Linear cyclodextrin copolymers
US7091192B1 (en) 1998-07-01 2006-08-15 California Institute Of Technology Linear cyclodextrin copolymers
US6245427B1 (en) 1998-07-06 2001-06-12 DüZGüNES NEJAT Non-ligand polypeptide and liposome complexes as intracellular delivery vehicles
US20040043489A1 (en) * 1998-07-08 2004-03-04 Menzo Havenga Gene delivery vectors provided with a tissue tropism for dendritic cells and methods of use
US7396919B1 (en) * 1998-07-17 2008-07-08 Mirus Bio Corporation Charge reversal of polyion complexes
US6077709A (en) 1998-09-29 2000-06-20 Isis Pharmaceuticals Inc. Antisense modulation of Survivin expression
US6333396B1 (en) 1998-10-20 2001-12-25 Enzon, Inc. Method for targeted delivery of nucleic acids
US20050063950A1 (en) * 1998-11-19 2005-03-24 Georgetown University Systemic viral/ligand gene delivery system and gene therapy
DE69942837D1 (de) 1998-11-19 2010-11-18 Univ Georgetown Systemisches virus/ligand genabgabemittel und gentherapie
US7098192B2 (en) 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
GB9908195D0 (en) 1999-04-09 1999-06-02 Microbiological Res Authority Treatment of intracellular infection
US6514762B1 (en) 1999-04-23 2003-02-04 New Mexico State University Technology Transfer Corporation Delivery of nucleotides by electrochemical release
DE19918446A1 (de) * 1999-04-23 2000-11-23 Univ Eberhard Karls Verwendung von Coxsackievirus B3 zur Verbesserung der Transfektion von Zellen
US20040009936A1 (en) * 1999-05-03 2004-01-15 Tang De-Chu C. Vaccine and drug delivery by topical application of vectors and vector extracts
US20050232900A1 (en) * 1999-05-18 2005-10-20 Crucell Holland B.V. Serotype of adenovirus and uses thereof
US6696272B1 (en) 1999-06-02 2004-02-24 Hsc Research & Development Limited Partnership Products and methods for gaucher disease therapy
DK1102785T3 (da) * 1999-06-07 2013-05-13 Arrowhead Res Corp Sammensætninger til lægemiddeltilførsel ved anvendelse af pH-følsomme molekyler
US20080281041A1 (en) * 1999-06-07 2008-11-13 Rozema David B Reversibly Masked Polymers
US8541548B2 (en) * 1999-06-07 2013-09-24 Arrowhead Madison Inc. Compounds and methods for reversible modification of biologically active molecules
ES2183752T3 (es) * 1999-09-17 2005-07-16 Tgt Laboratories, S.A. De C.V. Vectores adenovirales recombinantes y su utilizacion en el tratamiento de la cirrosis de higado.
CA2395636A1 (en) * 1999-12-30 2001-07-12 Novartis Ag Novel colloid synthetic vectors for gene therapy
AU2764801A (en) 2000-01-07 2001-07-24 University Of Washington Enhanced transport of agents using membrane disruptive agents
US6261840B1 (en) 2000-01-18 2001-07-17 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US20020055479A1 (en) 2000-01-18 2002-05-09 Cowsert Lex M. Antisense modulation of PTP1B expression
US6680172B1 (en) 2000-05-16 2004-01-20 Regents Of The University Of Michigan Treatments and markers for cancers of the central nervous system
EP1157999A1 (en) * 2000-05-24 2001-11-28 Introgene B.V. Methods and means for enhancing skin transplantation using gene delivery vehicles having tropism for primary fibroblasts, as well as other uses thereof
JP2003534806A (ja) * 2000-05-31 2003-11-25 ジェンベク、インコーポレイティッド アデノウイルスベクターのターゲティングの方法および組成物
JP2004501657A (ja) 2000-06-28 2004-01-22 マックス−デルブルック−セントラム フュール モレクラーレ メディツィン トランスフェクション効率の改良方法
US20040058856A1 (en) * 2000-07-26 2004-03-25 Soo-Young Choi Oligolysine transducing domain, oligolysine-cargo molecule complex and uses thereof
US20040033605A1 (en) * 2000-09-20 2004-02-19 Menzo Havenga Gene delivery vectors provided with a tissue tropism for dendritic cells
EP1191104A1 (en) * 2000-09-26 2002-03-27 Introgene B.V. Gene delivery vehicles and use thereof in the preparation of a medicament and/or vaccine
DE10049010A1 (de) * 2000-10-04 2002-04-18 Boehringer Ingelheim Int Transferrin-Polykation/DNS-Komplexe für die systemische Therapie von Tumorerkrankungen mit zytotoxischen Proteinen
US20020086849A1 (en) * 2000-10-27 2002-07-04 Gulilat Gebeyehu Method for introducing antisense oligonucleotides into eucaryotic cells
US6892140B1 (en) 2000-11-27 2005-05-10 Enteron, Inc. Immunogenic cancer peptides and uses thereof
MXPA00011713A (es) * 2000-11-28 2002-05-31 Tgt Lab S A De C V Vectores recombinantes virales y no virales conteniendo el gen humano del activador de plasminogeno derivado de urocinasa y su utilidad en el tratamiento de diversos tipos de fibrosis hepatica, renal, pulmonar, pancreatica, cardiaca y cicatrices hipe
US7635571B2 (en) * 2000-12-07 2009-12-22 Siemens Healthcare Diagnostics Products Gmbh Amplified signal in binding assays
US6635466B2 (en) * 2001-01-09 2003-10-21 University Of Iowa Research Foundation Adenovirus serotype 30 (Ad30)
US20030170826A1 (en) * 2001-02-02 2003-09-11 Peter Rabinovich Peptides for facilitating composite receptor expression and translocation of macromolecules
ES2310201T3 (es) 2001-02-13 2009-01-01 Government Of The United States Of America, As Represented By The Secretary Of The Army Vacuna para inmunizacion transcutanea contra la diarrea de los viajeros.
US20020150566A1 (en) * 2001-03-23 2002-10-17 Kun-Liang Guan Method of inhibiting cancerous cell proliferation using Ras mutants of GDP-bound conformation
US20020193295A1 (en) * 2001-05-04 2002-12-19 Emanuel Calenoff Immunogenic peptides and uses thereof
EP1383480A4 (en) * 2001-04-30 2006-05-24 Targeted Genetics Corp Lipid-Containing Drug Delivery Complexes and Method of Producing the Same
WO2002089586A1 (en) * 2001-05-10 2002-11-14 St. Jude Children's Research Hospital Lung epithelial cell line for propagating viruses
US20060159657A1 (en) * 2001-06-14 2006-07-20 Macromed, Incorporated Formulations of lymphokines and method of use thereof for local or both local and systemic control of proliferative cell disorders
US20030003074A1 (en) * 2001-06-14 2003-01-02 Macromed, Inc. Formulations of lymphokines and method of use thereof for local or both local and systemic control of proliferative cell disorders
US7803915B2 (en) 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
WO2003000113A2 (en) 2001-06-20 2003-01-03 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
EP2270024B1 (en) 2001-06-21 2018-10-24 Ionis Pharmaceuticals, Inc. Antisense modulation of superoxide dismutase 1, soluble expression
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
US6964950B2 (en) 2001-07-25 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
US20030096772A1 (en) 2001-07-30 2003-05-22 Crooke Rosanne M. Antisense modulation of acyl CoA cholesterol acyltransferase-2 expression
US7407943B2 (en) 2001-08-01 2008-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein B expression
US7227014B2 (en) 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
JP2005536439A (ja) 2001-09-18 2005-12-02 ジェネンテック・インコーポレーテッド 腫瘍の診断及び治療のための組成物と方法
WO2003052117A2 (en) * 2001-09-19 2003-06-26 Massachusetts Institute Of Technology Methods and products related to non-viral transfection
US6750019B2 (en) 2001-10-09 2004-06-15 Isis Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
NZ577565A (en) 2001-10-09 2010-10-29 Isis Pharmaceuticals Inc Antisense modulation of insulin-like growth factor binding protein 5 expressions
US7745418B2 (en) * 2001-10-12 2010-06-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting viral replication
WO2003088808A2 (en) 2002-04-16 2003-10-30 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
JP2005527639A (ja) 2001-11-02 2005-09-15 インサート セラピューティクス インコーポレイテッド Rna干渉の治療的利用のための方法及び組成物
US20030138407A1 (en) * 2001-11-02 2003-07-24 Patrick Lu Therapeutic methods for nucleic acid delivery vehicles
US6965025B2 (en) 2001-12-10 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
NZ556415A (en) 2002-01-02 2009-02-28 Genentech Inc Compositions and methods for the diagnosis and treatment of glioma tumor
US8138383B2 (en) * 2002-03-11 2012-03-20 Arrowhead Madison Inc. Membrane active heteropolymers
US8008355B2 (en) * 2002-03-11 2011-08-30 Roche Madison Inc. Endosomolytic poly(vinyl ether) polymers
US20030186916A1 (en) * 2002-03-12 2003-10-02 Lei Yu Vector for transfection of eukaryotic cells
AU2003225793A1 (en) * 2002-03-15 2003-09-29 Baxter Healthcare S.A. Methods and compositions for directing cells to target organs
US20030180712A1 (en) 2002-03-20 2003-09-25 Biostratum Ab Inhibition of the beta3 subunit of L-type Ca2+ channels
US20040142450A1 (en) * 2002-05-10 2004-07-22 Seo Sang Heui Lung epithelial cell line for propagating viruses
US7199107B2 (en) 2002-05-23 2007-04-03 Isis Pharmaceuticals, Inc. Antisense modulation of kinesin-like 1 expression
US20050233993A1 (en) * 2002-06-05 2005-10-20 Kadonaga James T Methods for promoting homologous recombination
PT2332582E (pt) 2002-06-19 2014-01-28 Apeiron Biologics Ag Activação de ace2 para o tratamento de doença cardíaca, pulmonar e renal e de hipertensão
US20060003316A1 (en) * 2002-07-15 2006-01-05 John Simard Immunogenic compositions derived from poxviruses and methods of using same
PT1534340E (pt) 2002-09-06 2012-03-13 Cerulean Pharma Inc Polímeros à base de ciclodextrina para a administração de medicamentos ligados por ligação covalente
CN1820078A (zh) * 2002-09-09 2006-08-16 田纳西大学研究基金会 弹状病毒的重组突变体及其应用方法
MXPA05002791A (es) 2002-09-13 2005-12-05 Replicor Inc Oligonucleotidos antivirales complementarios sin secuencia.
AU2003278957A1 (en) 2002-09-26 2004-04-23 Amgen, Inc. Modulation of forkhead box o1a expression
MXPA05003843A (es) * 2002-10-10 2006-02-17 Us Dept Veterans Affairs Deteccion, localizacion y determicnacion de fases de tumores usando linfocitos activados rotulados dirigidos a un epitope especifico.
AU2003295387A1 (en) 2002-11-05 2004-06-03 Isis Parmaceuticals, Inc. Modified oligonucleotides for use in rna interference
EP1560839A4 (en) 2002-11-05 2008-04-23 Isis Pharmaceuticals Inc CHIMERIC OLIGOMER COMPOUNDS AND THEIR USE IN GENE MODULATION
US20080026077A1 (en) * 2002-11-12 2008-01-31 John Hilfinger Methods and compositions of gene delivery agents for systemic and local therapy
US20050026859A1 (en) * 2002-11-12 2005-02-03 John Hilfinger Methods and compositions of gene delivery agents for systemic and local therapy
ES2420914T3 (es) 2002-11-13 2013-08-27 Genzyme Corporation Modulación antisentido de la expresión de la apolipoproteína B
CA2505801A1 (en) 2002-11-13 2004-05-27 Rosanne Crooke Antisense modulation of apolipoprotein b expression
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
US7468356B2 (en) 2003-02-11 2008-12-23 Antisense Therapeutics Ltd. Modulation of insulin like growth factor I receptor expression
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
US20040185559A1 (en) 2003-03-21 2004-09-23 Isis Pharmaceuticals Inc. Modulation of diacylglycerol acyltransferase 1 expression
US7217566B2 (en) * 2003-03-24 2007-05-15 Invitrogen Corporation Attached cell lines
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
EP1620112A4 (en) * 2003-04-17 2007-04-25 Univ Columbia DESMOGLEIN 4 IS A NEW GENUS INVOLVED IN HAIR GROWTH
US7399853B2 (en) 2003-04-28 2008-07-15 Isis Pharmaceuticals Modulation of glucagon receptor expression
US20040220085A1 (en) * 2003-05-02 2004-11-04 Jasbir Sandhu Compositions for nucleic acid delivery
US20040220084A1 (en) * 2003-05-02 2004-11-04 Jasbir Sandhu Methods for nucleic acid delivery
BRPI0410886A (pt) 2003-06-03 2006-07-04 Isis Pharmaceuticals Inc composto de filamento duplo, composição farmacêutica, sal farmaceuticamente aceitável, métodos de modificação do ácido nucleico que codifica a survivina humana, de inibição da expressão da suvivina em células ou tecidos, e de tratamento de uma condição associada com a expressão ou superexpressão da suvivina, e, oligonucleotìdeo de rnai de filamento único
CA2528094A1 (en) * 2003-06-09 2005-01-13 Corixa Corporation Expression vectors for stimulating an immune response to cancer antigens
US7883720B2 (en) * 2003-07-09 2011-02-08 Wisconsin Alumni Research Foundation Charge-dynamic polymers and delivery of anionic compounds
WO2005013901A2 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
NZ576775A (en) 2003-09-18 2010-12-24 Isis Pharmaceuticals Inc Modulation of eIF4E expression
NZ546272A (en) 2003-10-10 2009-05-31 Alchemia Oncology Pty Ltd The modulation of hyaluronan synthesis and degradation in the treatment of disease
US8062891B2 (en) 2003-10-24 2011-11-22 Gencia Corporation Nonviral vectors for delivering polynucleotides to plants
US8507277B2 (en) 2003-10-24 2013-08-13 Gencia Corporation Nonviral vectors for delivering polynucleotides
US20090123468A1 (en) 2003-10-24 2009-05-14 Gencia Corporation Transducible polypeptides for modifying metabolism
US20090208478A1 (en) * 2003-10-24 2009-08-20 Gencia Corporation Transducible polypeptides for modifying metabolism
US8133733B2 (en) 2003-10-24 2012-03-13 Gencia Corporation Nonviral vectors for delivering polynucleotides to target tissues
EP2418281B1 (en) 2003-10-24 2016-06-01 Gencia Corporation Methods and compositions for delivering polynucleotides
US20050191653A1 (en) 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
CA2747871C (en) 2003-11-17 2018-04-10 Genentech, Inc. Compositions and methods for the treatment of tumor of hematopoietic origin
US20050265956A1 (en) * 2004-01-12 2005-12-01 Ye Liu Polyalkyleneimine-graft-biodegradable polymers for delivery of bioactive agents
US20050164271A1 (en) 2004-01-20 2005-07-28 Sanjay Bhanot Modulation of glucocorticoid receptor expression
US7468431B2 (en) 2004-01-22 2008-12-23 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
WO2005069987A2 (en) * 2004-01-23 2005-08-04 City Of Hope Amplifying interfering rna (rnai) expression and effects
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
US20050203047A1 (en) * 2004-03-10 2005-09-15 Ulrich Thomann Delivery vectors for short interfering RNA, micro-RNA and antisense RNA
EP2700720A3 (en) 2004-03-15 2015-01-28 Isis Pharmaceuticals, Inc. Compositions and methods for optimizing cleavage of RNA by RNASE H
WO2005097207A2 (en) * 2004-03-26 2005-10-20 Curis, Inc. Rna interference modulators of hedgehog signaling and uses thereof
US20050244869A1 (en) 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
US8697139B2 (en) 2004-09-21 2014-04-15 Frank M. Phillips Method of intervertebral disc treatment using articular chondrocyte cells
US9315862B2 (en) 2004-10-05 2016-04-19 California Institute Of Technology Aptamer regulated nucleic acids and uses thereof
US7833513B2 (en) 2004-12-03 2010-11-16 Rhode Island Hospital Treatment of Alzheimer's Disease
ES2852549T3 (es) 2005-02-09 2021-09-13 Sarepta Therapeutics Inc Composición antisentido para tratamiento de la atrofia muscular
JP2008531581A (ja) * 2005-02-23 2008-08-14 ユーエービー リサーチ ファウンデーション アルキル−グリコシドで増強されたワクチン接種
CN101175769A (zh) 2005-03-10 2008-05-07 健泰科生物技术公司 用于调控血管完整性的方法和组合物
AU2006230436B2 (en) 2005-03-31 2011-11-24 Calando Pharmaceuticals, Inc. Inhibitors of ribonucleotide reductase subunit 2 and uses thereof
US8734851B2 (en) * 2005-04-29 2014-05-27 Wisconsin Alumni Research Foundation Localized delivery of nucleic acid by polyelectrolyte assemblies
AU2006318194B2 (en) 2005-11-21 2012-08-09 Isis Pharmaceuticals, Inc. Modulation of eiF4E-BP2 expression
US20090317802A1 (en) * 2005-12-09 2009-12-24 Bhatia Sangeeta N Compositions and Methods to Monitor RNA Delivery to Cells
CA2566267A1 (en) * 2005-12-09 2007-06-09 University Health Network Thymidylate kinase mutants and uses thereof
US20090068158A1 (en) * 2005-12-09 2009-03-12 Medin Jeffrey A Thymidylate kinase mutants and uses thereof
WO2007089584A2 (en) 2006-01-26 2007-08-09 Isis Pharmaceuticals, Inc. Compositions and their uses directed to huntingtin
CA2638915A1 (en) * 2006-02-10 2007-08-23 The Regents Of The University Of California Transducible delivery of sirna by dsrna binding domain fusions to ptd/cpps
EP2502999A3 (en) 2006-03-03 2013-01-23 Amorfix Life Sciences Ltd. Methods and compositions to treat and detect misfolded-SOD1 mediated diseases
US7718193B2 (en) * 2006-03-16 2010-05-18 University Of Washington Temperature- and pH-responsive polymer compositions
EP2614839A3 (en) 2006-04-05 2015-01-28 Genentech, Inc. Method for using BOC/CDO to modulate hedgehog signaling
MX2008014005A (es) 2006-05-03 2009-01-27 Baltic Technology Dev Ltd Agentes antisentido que combinan un oligonucleotido modificado con base fuertemente unida y nucleasa artificial.
JP6125741B2 (ja) * 2006-07-12 2017-05-17 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 可逆的なホスホトリエステル電荷中和保護基による核酸の形質導入可能な送達
WO2008011473A2 (en) 2006-07-19 2008-01-24 Isis Pharmaceuticals, Inc. Compositions and their uses directed to hbxip
CN102614528B (zh) * 2006-08-18 2014-02-26 箭头研究公司 用于体内递送多核苷酸的多缀合物
US8017109B2 (en) * 2006-08-18 2011-09-13 Roche Madison Inc. Endosomolytic poly(acrylate) polymers
KR20090066289A (ko) 2006-09-08 2009-06-23 로드아일랜드하스피틀 알코올 유발성 뇌 질환의 치료, 예방 및 역행
EP2061484B1 (en) 2006-09-08 2012-11-07 Rhode Island Hospital Treatment, prevention, and reversal of alcohol-induced liver disease
WO2008058291A2 (en) 2006-11-09 2008-05-15 California Institute Of Technology Modular aptamer-regulated ribozymes
AU2007333225B2 (en) * 2006-12-08 2014-06-12 Massachusetts Institute Of Technology Delivery of nanoparticles and/or agents to cells
US8048998B2 (en) * 2007-01-19 2011-11-01 Exiqon A/S Mediated cellular delivery of LNA oligonucleotides
US8834918B2 (en) * 2007-01-22 2014-09-16 Wisconsin Alumni Research Foundation Modified multilayered film
JP2010516625A (ja) 2007-01-24 2010-05-20 インサート セラピューティクス, インコーポレイテッド 制御された薬物送達のためのテザー基を有するポリマー−薬物コンジュゲート
EP2114981B1 (en) 2007-01-29 2013-05-08 Isis Pharmaceuticals, Inc. Compounds and methods for modulating protein expression
JP5761915B2 (ja) 2007-02-22 2015-08-12 ジェネンテック, インコーポレイテッド 炎症性腸疾患の検出方法
US7981688B2 (en) 2007-03-08 2011-07-19 University Of Washington Stimuli-responsive magnetic nanoparticles and related methods
CA2584494A1 (en) * 2007-03-27 2008-09-27 Jeffrey A. Medin Vector encoding therapeutic polypeptide and safety elements to clear transduced cells
ES2654303T3 (es) 2007-05-04 2018-02-13 University Health Network Inmunoterapia de IL-12 contra el cáncer
US20090082217A1 (en) * 2007-07-16 2009-03-26 California Institute Of Technology Selection of nucleic acid-based sensor domains within nucleic acid switch platform
US20120165387A1 (en) 2007-08-28 2012-06-28 Smolke Christina D General composition framework for ligand-controlled RNA regulatory systems
US8367815B2 (en) * 2007-08-28 2013-02-05 California Institute Of Technology Modular polynucleotides for ligand-controlled regulatory systems
US8865667B2 (en) * 2007-09-12 2014-10-21 California Institute Of Technology Higher-order cellular information processing devices
AU2008307482B2 (en) 2007-10-02 2012-07-12 Amgen Inc. Increasing erythropoietin using nucleic acids hybridizable to micro-RNA and precursors thereof
US20090105375A1 (en) * 2007-10-09 2009-04-23 Lynn David M Ultrathin Multilayered Films for Controlled Release of Anionic Reagents
US9029524B2 (en) * 2007-12-10 2015-05-12 California Institute Of Technology Signal activated RNA interference
EP2077119A1 (de) * 2007-12-21 2009-07-08 Apeiron Biologics Forschungs- und Entwicklungsgesellschaft M.B.H. Behandlung von Fibrosen und Lebererkrankungen
US8344116B2 (en) * 2008-03-17 2013-01-01 Case Western Reserve University Polymers and complexes for delivery of nucleic acids to intracellular targets
US8568709B2 (en) 2008-03-20 2013-10-29 University Health Network Thymidylate kinase fusions and uses thereof
CN102037123A (zh) 2008-04-04 2011-04-27 卡兰多制药股份有限公司 Epas1抑制剂的组合物和用途
GB2459436A (en) * 2008-04-08 2009-10-28 Henderson Morley Plc Vaccine adjuvant
US8324333B2 (en) 2008-06-05 2012-12-04 Wisconsin Alumni Research Foundation Anionic charge-dynamic polymers for release of cationic agents
US8815818B2 (en) 2008-07-18 2014-08-26 Rxi Pharmaceuticals Corporation Phagocytic cell delivery of RNAI
CN103429270B (zh) 2008-08-25 2016-11-23 埃克斯雷德制药有限公司 阻止结缔组织生长因子的反义核苷酸及其用途
CA2746527A1 (en) 2008-09-22 2010-03-25 Rxi Pharmaceuticals Corporation Rna interference in skin indications
CA2745811C (en) 2008-12-04 2021-07-13 Joseph Collard Treatment of tumor suppressor gene related diseases by inhibition of natural antisense transcript to the gene
JP6091752B2 (ja) 2008-12-04 2017-03-08 クルナ・インコーポレーテッド Epoに対する天然アンチセンス転写物の抑制によるエリスロポエチン(epo)関連疾患の治療
MX366774B (es) 2008-12-04 2019-07-24 Curna Inc Uso de oligonucleótidos antisentido en la inhibición de transcrito antisentido natural para sirtuina 1.
US9493774B2 (en) 2009-01-05 2016-11-15 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAi
JP6066035B2 (ja) 2009-02-12 2017-01-25 クルナ・インコーポレーテッド グリア細胞由来神経栄養因子(gdnf)関連疾病の、gdnfに対する天然アンチセンス転写物の抑制による治療
US9074210B2 (en) 2009-02-12 2015-07-07 Curna, Inc. Treatment of brain derived neurotrophic factor (BDNF) related diseases by inhibition of natural antisense transcript to BDNF
US8329882B2 (en) 2009-02-18 2012-12-11 California Institute Of Technology Genetic control of mammalian cells with synthetic RNA regulatory systems
EP2963116B1 (en) 2009-03-04 2020-11-11 CuRNA, Inc. Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirt 1
US9464287B2 (en) 2009-03-16 2016-10-11 Curna, Inc. Treatment of nuclear factor (erythroid-derived 2)-like 2 (NRF2) related diseases by inhibition of natural antisense transcript to NRF2
MX2011009752A (es) 2009-03-17 2011-09-29 Opko Curna Llc Tratamiento de enfermedades relacionadas a homologo tipo delta 1(dlk1) por inhibicion de transcrito antisentido natural a homologo tipo delta (dlk1).
US9145555B2 (en) 2009-04-02 2015-09-29 California Institute Of Technology Integrated—ligand-responsive microRNAs
EP3248618A1 (en) 2009-04-22 2017-11-29 Massachusetts Institute Of Technology Innate immune suppression enables repeated delivery of long rna molecules
CN102639151B (zh) 2009-05-01 2017-03-22 库尔纳公司 通过针对hbf/hbg的天然反义转录物的抑制治疗血红蛋白(hbf/hbg)相关疾病
WO2010129746A2 (en) 2009-05-06 2010-11-11 Curna, Inc. Treatment of tristetraproline (ttp) related diseases by inhibition of natural antisense transcript to ttp
JP5883782B2 (ja) 2009-05-06 2016-03-15 クルナ・インコーポレーテッド 脂質輸送代謝遺伝子に対する天然アンチセンス転写物の抑制による脂質輸送代謝遺伝子関連疾患の治療
CA2762369C (en) 2009-05-18 2021-12-28 Joseph Collard Treatment of reprogramming factor related diseases by inhibition of natural antisense transcript to a reprogramming factor
EP2432882B1 (en) 2009-05-22 2019-12-25 CuRNA, Inc. TREATMENT OF TRANSCRIPTION FACTOR E3 (TFE3) and INSULIN RECEPTOR SUBSTRATE 2 (IRS2) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO TFE3
CN103221541B (zh) 2009-05-28 2017-03-01 库尔纳公司 通过抑制抗病毒基因的天然反义转录物来治疗抗病毒基因相关疾病
US8426214B2 (en) * 2009-06-12 2013-04-23 University Of Washington System and method for magnetically concentrating and detecting biomarkers
CA2765700C (en) 2009-06-16 2021-01-05 Opko Curna, Llc Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene
ES2629339T3 (es) 2009-06-16 2017-08-08 Curna, Inc. Tratamiento de enfermedades relacionadas con la paraoxonasa 1 (pon1) por inhibición de transcrito antisentido natural a pon1
US8859515B2 (en) 2009-06-24 2014-10-14 Curna, Inc. Treatment of tumor necrosis factor receptor 2 (TNFR2) related diseases by inhibition of natural antisense transcript to TNFR2
CN102482672B (zh) 2009-06-26 2016-11-09 库尔纳公司 通过抑制唐氏综合征基因的天然反义转录物治疗唐氏综合征基因相关疾病
US9234199B2 (en) 2009-08-05 2016-01-12 Curna, Inc. Treatment of insulin gene (INS) related diseases by inhibition of natural antisense transcript to an insulin gene (INS)
CA2771172C (en) 2009-08-25 2021-11-30 Opko Curna, Llc Treatment of 'iq motif containing gtpase activating protein' (iqgap) related diseases by inhibition of natural antisense transcript to iqgap
JP5887270B2 (ja) 2009-09-02 2016-03-16 ジェネンテック, インコーポレイテッド 突然変異体smoothenedおよびその使用方法
US20130079382A1 (en) 2009-10-12 2013-03-28 Larry J. Smith Methods and Compositions for Modulating Gene Expression Using Oligonucleotide Based Drugs Administered in vivo or in vitro
WO2011050194A1 (en) 2009-10-22 2011-04-28 Genentech, Inc. Methods and compositions for modulating hepsin activation of macrophage-stimulating protein
WO2011052804A1 (en) * 2009-10-30 2011-05-05 Tokyo University Of Science Educational Foundation Administrative Organization Method of delivering agent into target cell
US20120244169A1 (en) 2009-11-06 2012-09-27 Fibrogen, Inc. Treatment for Radiation-Induced Disorders
US9080933B2 (en) 2009-11-09 2015-07-14 University Of Washington Through Its Center For Commercialization Stimuli-responsive polymer diagnostic assay comprising magnetic nanoparticles and capture conjugates
US20110117668A1 (en) * 2009-11-09 2011-05-19 University Of Washington Through Its Center For Commercialization Self-powered smart diagnostic devices
JP6220126B2 (ja) * 2009-11-23 2017-10-25 セルリアン・ファーマ・インコーポレイテッド 治療的送達のためのシクロデキストリンに基づく重合体
MX2012006072A (es) 2009-11-30 2012-07-23 Genentech Inc Composiciones y metodos para el diagnostico y el tratamiento de tumores.
AU2010329847A1 (en) 2009-12-11 2012-07-26 Genecode As Methods of facilitating neural cell survival using GDNF family ligand (GFL) mimetics or RET signaling pathway activators
JP6025567B2 (ja) 2009-12-16 2016-11-16 カッパーアールエヌエー,インコーポレイテッド 膜結合転写因子ペプチダーゼ、部位1(mbtps1)に対する天然アンチセンス転写物の阻害によるmbtps1関連性疾患の治療
WO2011079263A2 (en) 2009-12-23 2011-06-30 Curna, Inc. Treatment of uncoupling protein 2 (ucp2) related diseases by inhibition of natural antisense transcript to ucp2
US8940708B2 (en) 2009-12-23 2015-01-27 Curna, Inc. Treatment of hepatocyte growth factor (HGF) related diseases by inhibition of natural antisense transcript to HGF
RU2611186C2 (ru) 2009-12-29 2017-02-21 Курна, Инк. ЛЕЧЕНИЕ ЗАБОЛЕВАНИЙ, СВЯЗАННЫХ С ОПУХОЛЕВЫМ БЕЛКОМ 63 (р63), ПУТЕМ ИНГИБИРОВАНИЯ ПРИРОДНОГО АНТИСМЫСЛОВОГО ТРАНСКРИПТА К р63
RU2615450C2 (ru) 2009-12-29 2017-04-04 Курна, Инк. Лечение заболеваний, связанных с ядерным респираторным фактором 1(nrf1), путем ингибирования природного антисмыслового транскрипта к nrf1
US8946181B2 (en) 2010-01-04 2015-02-03 Curna, Inc. Treatment of interferon regulatory factor 8 (IRF8) related diseases by inhibition of natural antisense transcript to IRF8
WO2011085066A2 (en) 2010-01-06 2011-07-14 Curna, Inc. Treatment of pancreatic developmental gene related diseases by inhibition of natural antisense transcript to a pancreatic developmental gene
CA2786535C (en) 2010-01-11 2019-03-26 Curna, Inc. Treatment of sex hormone binding globulin (shbg) related diseases by inhibition of natural antisense transcript to shbg
WO2011090971A2 (en) 2010-01-19 2011-07-28 The Trustees Of Columbia University In The City Of New York Osteocalcin as a treatment for male reproductive disorders
RU2611192C2 (ru) 2010-01-25 2017-02-21 Курна, Инк. ЛЕЧЕНИЕ ЗАБОЛЕВАНИЙ, СВЯЗАННЫХ С РНКазой Н1, ПУТЕМ ИНГИБИРОВАНИЯ ПРИРОДНОГО АНТИСМЫСЛОВОГО ТРАНСКРИПТА К РНКазе Н1
EP2539452B1 (en) 2010-02-22 2016-07-27 CuRNA, Inc. Treatment of pyrroline-5-carboxylate reductase 1 (pycr1) related diseases by inhibition of natural antisense transcript to pycr1
KR20130004579A (ko) 2010-02-23 2013-01-11 제넨테크, 인크. 종양의 진단 및 치료를 위한 조성물 및 방법
SI2539451T1 (sl) 2010-02-24 2016-04-29 Arrowhead Research Corporation Sestavki za ciljano dostavo sirna
EP2556160A4 (en) 2010-04-09 2013-08-21 Curna Inc TREATMENT OF ILLNESSES ASSOCIATED WITH FIBROBLASTIC GROWTH FACTOR 21 (FGF21) BY INHIBITION OF THE NATURAL ANTISENSE TRANSCRIPT AGAINST FGF21
US8362207B2 (en) 2010-04-16 2013-01-29 Wake Forest University Health Sciences Multi-level specific targeting of cancer cells with IL-13
DK2563920T3 (en) 2010-04-29 2017-05-22 Ionis Pharmaceuticals Inc Modulation of transthyretin expression
BR112012028010A2 (pt) 2010-05-03 2017-09-26 Genentech Inc anticorpo isolado, célula, ácido nucleíco isolado, método de identificação de um primeiro anticorpo que se liga a um epítopo antigênico tat425 ligado or um anticorpo, métodos de inibir o crescimento de uma célula, de tratamento terapêutico de determinação da presença de uma proteína de tat425 e de diagnóstico da presença de um tumor em um mamífero
RU2018110642A (ru) 2010-05-03 2019-02-27 Курна, Инк. Лечение заболеваний, связанных с сиртуином (sirt), путем ингибирования природного антисмыслового транскрипта к сиртуину (sirt)
EP2569430B1 (en) 2010-05-12 2018-10-17 The Trustees of Columbia University in the City of New York Methods for producing enteroendocrine cells that make and secrete insulin
TWI531370B (zh) 2010-05-14 2016-05-01 可娜公司 藉由抑制par4天然反股轉錄本治療par4相關疾病
EP3299464B1 (en) 2010-05-26 2019-10-02 CuRNA, Inc. Treatment of atonal homolog 1 (atoh1) related diseases by inhibition of natural antisense transcript to atoh1
US9057067B2 (en) 2010-07-10 2015-06-16 Kinki University Method for transfecting nucleic acid to cell and nucleic acid complex
NO2593547T3 (sl) 2010-07-14 2018-04-14
DK2625197T3 (en) 2010-10-05 2016-10-03 Genentech Inc Smoothened MUTANT AND METHODS OF USING THE SAME
CA2813901C (en) 2010-10-06 2019-11-12 Curna, Inc. Treatment of sialidase 4 (neu4) related diseases by inhibition of natural antisense transcript to neu4
US9222088B2 (en) 2010-10-22 2015-12-29 Curna, Inc. Treatment of alpha-L-iduronidase (IDUA) related diseases by inhibition of natural antisense transcript to IDUA
DK2633052T3 (en) 2010-10-27 2018-07-16 Curna Inc TREATMENT OF INTERFERON-RELATED DEVELOPMENT REGULATOR 1 (IFRD1) -RELATED DISEASES BY INHIBITION OF NATURAL ANTISENCE TRANSCRIPT TO IFRD1
WO2012061811A2 (en) 2010-11-05 2012-05-10 Fibrogen, Inc. Treatment method for lung remodeling diseases
CN103459599B (zh) 2010-11-23 2017-06-16 库尔纳公司 通过抑制nanog的天然反义转录物而治疗nanog相关疾病
SI2670411T1 (sl) 2011-02-02 2019-06-28 Excaliard Pharmaceuticals, Inc. Protismiselne spojine, ki so usmerjene na rastni faktor veznega tkiva (CTGF), za uporabo v postopku zdravljenja keloidov ali hipertrofnih brazgotin
WO2012109495A1 (en) 2011-02-09 2012-08-16 Metabolic Solutions Development Company, Llc Cellular targets of thiazolidinediones
WO2012124688A1 (ja) 2011-03-14 2012-09-20 国立大学法人北海道大学 肺送達のためのベクター、導入剤及び使用
US8658783B2 (en) 2011-04-13 2014-02-25 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
RU2620980C2 (ru) 2011-06-09 2017-05-30 Курна, Инк. Лечение заболеваний, связанных с фратаксином (fxn), путем ингибирования природного антисмыслового транскрипта fxn
AU2012271357A1 (en) 2011-06-16 2013-05-02 Ionis Pharmaceuticals, Inc. Antisense modulation of fibroblast growth factor receptor 4 expression
AU2012312433B2 (en) 2011-09-20 2017-07-06 Ionis Pharmaceuticals, Inc. Antisense modulation of GCGR expression
US20130085139A1 (en) 2011-10-04 2013-04-04 Royal Holloway And Bedford New College Oligomers
EP2771463A4 (en) 2011-10-25 2015-09-09 Isis Pharmaceuticals Inc ANTISENSE MODULATION OF GCCR EXPRESSION
JP2015511494A (ja) 2012-03-15 2015-04-20 キュアナ,インク. 脳由来神経栄養因子(bdnf)に対する天然アンチセンス転写物の阻害によるbdnf関連の疾患の処置
US9950001B2 (en) 2012-08-20 2018-04-24 The Regents Of The University Of California Polynucleotides having bioreversible groups
EP2943194A1 (en) 2012-09-17 2015-11-18 Chemedest Ltd. Treatment of peripheral neuropathy using gfr(alpha)3 type receptor agonists
WO2014055493A1 (en) 2012-10-02 2014-04-10 Cerulean Pharma Inc. Methods and systems for polymer precipitation and generation of particles
US10415024B2 (en) 2012-11-16 2019-09-17 Poseida Therapeutics, Inc. Site-specific enzymes and methods of use
US10052364B2 (en) 2013-03-15 2018-08-21 The Trustees Of Columbia University In The City Of New York Osteocalcin as a treatment for cognitive disorders
WO2014197938A1 (en) 2013-06-13 2014-12-18 Antisense Therapeutics Ltd Combination therapy
AU2014317961B2 (en) 2013-09-05 2020-07-30 Murdoch University Antisense-induced exon2 inclusion in acid alpha-glucosidase
EP3047023B1 (en) 2013-09-19 2019-09-04 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Compositions and methods for inhibiting jc virus (jcv)
US9975942B2 (en) 2013-11-11 2018-05-22 Wake Forest University Health Services EPHA3 And multi-valent targeting of tumors
WO2015116902A1 (en) 2014-01-31 2015-08-06 Genentech, Inc. G-protein coupled receptors in hedgehog signaling
WO2015120075A2 (en) 2014-02-04 2015-08-13 Genentech, Inc. Mutant smoothened and methods of using the same
WO2015171918A2 (en) 2014-05-07 2015-11-12 Louisiana State University And Agricultural And Mechanical College Compositions and uses for treatment thereof
EP3145550B1 (en) * 2014-05-19 2019-03-27 BioNTech AG Particles comprising protamine and rna in combination with endosome destabilizing agents
US10487314B2 (en) 2014-06-26 2019-11-26 The Trustees Of Columbia University In The City Of New York Inhibition of serotonin expression in gut enteroendocrine cells results in conversion to insulin-positive cells
WO2016033424A1 (en) 2014-08-29 2016-03-03 Genzyme Corporation Methods for the prevention and treatment of major adverse cardiovascular events using compounds that modulate apolipoprotein b
EP3226889A4 (en) 2014-11-19 2018-11-21 The Trustees of Columbia University in the City of New York Osteocalcin as a treatment for frailty associated with aging
JP2018504380A (ja) 2014-12-18 2018-02-15 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Reversir(商標)化合物
MA41795A (fr) 2015-03-18 2018-01-23 Sarepta Therapeutics Inc Exclusion d'un exon induite par des composés antisens dans la myostatine
EP3302497A4 (en) 2015-06-01 2019-01-16 Sarepta Therapeutics, Inc. EXCLUSION OF EXON INDUCED PAT TECHNOLOGY ANTISENSE IN COLLAGEN TYPE VII
DK3310909T3 (da) 2015-06-17 2021-09-13 Poseida Therapeutics Inc Sammensætninger og fremgangsmåder til at føre proteiner til specifikke loci i genomet
US10954300B2 (en) 2015-09-28 2021-03-23 The Trustees Of Columbia University In The City Of New York Use of pentoxifylline with immune checkpoint-blockade therapies for the treatment of melanoma
EP3359668A4 (en) 2015-10-09 2019-06-05 Sarepta Therapeutics, Inc. COMPOSITIONS AND METHODS OF TREATING DUCHENNE MUSCLE DYSTROPHY AND ASSOCIATED ILLNESSES THEREOF
EP3370734B1 (en) 2015-11-05 2023-01-04 Children's Hospital Los Angeles Antisense oligo for use in treating acute myeloid leukemia
EP3411396A1 (en) 2016-02-04 2018-12-12 Curis, Inc. Mutant smoothened and methods of using the same
EP3445405A4 (en) 2016-04-18 2019-12-18 Sarepta Therapeutics, Inc. ANTISENSE OLIGOMERS AND METHOD FOR USE THEREOF TO TREAT DISEASES RELATING TO THE ACID ALPHA GLUCOSIDASE GENE
JP2019532027A (ja) 2016-08-17 2019-11-07 ソルスティス バイオロジクス,リミティッド ポリヌクレオチド構築物
US10994025B2 (en) 2017-05-12 2021-05-04 Massachusetts Institute Of Technology Argonaute protein-double stranded RNA complexes and uses related thereto
WO2019006455A1 (en) 2017-06-30 2019-01-03 Solstice Biologics, Ltd. AUXILIARIES OF CHIRAL PHOSPHORAMIDITIS AND METHODS OF USE THEREOF
US11555189B2 (en) 2017-10-18 2023-01-17 Sarepta Therapeutics, Inc. Antisense oligomer compounds
WO2019126578A1 (en) 2017-12-20 2019-06-27 Poseida Therapeutics, Inc. Compositions and methods for directing proteins to specific loci in the genome
AU2019247490A1 (en) 2018-04-06 2020-10-22 Children's Medical Center Corporation Compositions and methods for somatic cell reprogramming and modulating imprinting
JP2024516168A (ja) 2021-04-22 2024-04-12 デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド がんを治療するための組成物および方法

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0388758A1 (de) * 1989-03-16 1990-09-26 BOEHRINGER INGELHEIM INTERNATIONAL GmbH Neue Protein-Polykation-Konjugate
EP0422543A1 (en) * 1989-10-09 1991-04-17 The Green Cross Corporation pH-sensitive liposomes
WO1991017773A2 (de) * 1990-05-18 1991-11-28 Boehringer Ingelheim International Gmbh Neue protein-polykation-konjugate

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IN165717B (sl) * 1986-08-07 1989-12-23 Battelle Memorial Institute
US5166320A (en) * 1987-04-22 1992-11-24 University Of Connecticut Carrier system and method for the introduction of genes into mammalian cells
US5428132A (en) * 1987-10-11 1995-06-27 United States Of America Conjugate and method for integration of foreign DNA into cells
US5192553A (en) * 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US5149782A (en) * 1988-08-19 1992-09-22 Tanox Biosystems, Inc. Molecular conjugates containing cell membrane-blending agents
US5108921A (en) * 1989-04-03 1992-04-28 Purdue Research Foundation Method for enhanced transmembrane transport of exogenous molecules
US5240846A (en) * 1989-08-22 1993-08-31 The Regents Of The University Of Michigan Gene therapy vector for cystic fibrosis
AU660629B2 (en) * 1990-10-01 1995-07-06 University Of Connecticut, The Targeting viruses and cells for selective internalization by cells
WO1992019749A1 (en) * 1991-05-03 1992-11-12 The Board Of Regents Acting For And On Behalf Of The University Of Michigan Targeted delivery of genes encoding cell surface receptors
JPH06510524A (ja) * 1991-05-14 1994-11-24 ユニバーシティ オブ コネチカット 免疫原性タンパク質をコードする遺伝子の標的への配達
DE69231385T2 (de) * 1991-06-05 2001-04-12 Univ Connecticut Storrs Zielgerichtete freisetzung von genen, die sekretorische proteine kodieren
EP0666923A1 (en) * 1991-09-05 1995-08-16 The University Of Connecticut Targeted delivery of poly- or oligonucleotides to cells
NZ244306A (en) 1991-09-30 1995-07-26 Boehringer Ingelheim Int Composition for introducing nucleic acid complexes into eucaryotic cells, complex containing nucleic acid and endosomolytic agent, peptide with endosomolytic domain and nucleic acid binding domain and preparation
US5521291A (en) * 1991-09-30 1996-05-28 Boehringer Ingelheim International, Gmbh Conjugates for introducing nucleic acid into higher eucaryotic cells
US5981273A (en) 1991-09-30 1999-11-09 Boehringer Ingelheim Int'l. Gmbh Composition comprising an endosomolytic agent for introducing nucleic acid complexes into higher eucaryotic cells
US5225182A (en) * 1991-10-31 1993-07-06 Sharma Yash P Vectored drug delivery system using a cephaloplastin linking agent and a methed of using the system
US5922859A (en) 1992-02-01 1999-07-13 Boehringer Ingelheim International Gmbh Complexes containing nucleic acid which can be taken-up by endocytosis into higher eukaryotic cells
US5583020A (en) * 1992-11-24 1996-12-10 Ribozyme Pharmaceuticals, Inc. Permeability enhancers for negatively charged polynucleotides
WO1994017832A1 (en) 1993-02-09 1994-08-18 The Scripps Research Institute Targeting and delivery of genes and antiviral agents into cells by the adenovirus penton
DE4311651A1 (de) 1993-04-08 1994-10-13 Boehringer Ingelheim Int Virus für den Transport von Fremd-DNA in höhere eukaryotische Zellen
DE4335025A1 (de) 1993-10-14 1995-04-20 Boehringer Ingelheim Int Endosomolytisch wirksame Partikel
US5928944A (en) 1994-02-04 1999-07-27 The United States Of America As Represented By The Department Of Health And Human Services Method of adenoviral-medicated cell transfection

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0388758A1 (de) * 1989-03-16 1990-09-26 BOEHRINGER INGELHEIM INTERNATIONAL GmbH Neue Protein-Polykation-Konjugate
EP0422543A1 (en) * 1989-10-09 1991-04-17 The Green Cross Corporation pH-sensitive liposomes
WO1991017773A2 (de) * 1990-05-18 1991-11-28 Boehringer Ingelheim International Gmbh Neue protein-polykation-konjugate

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
GENE Bd. 63, Nr. 2, 1988, AMSTERDAM NL Seiten 321 - 330 TIKCHONENKO, T.I. ET AL. 'Transfer of condensed viral DNA into eukaryotic cells using proteoliposomes' *
GENE Bd. 84, Nr. 2, 14. Dezember 1989, AMSTERDAM NL Seiten 429 - 438 GOULD-FOGERITE, S. ET AL. 'Chimerasome-mediated gene transfer in vitro and in vivo' *
JOURNAL OF GENERAL VIROLOGY Bd. 69, Nr. 8, August 1988, UK Seiten 1847 - 1857 WHARTON, S.A. ET AL. 'Membrane fusion by peptide analogues of Influenza Virus haemagglutinin' *
JOURNAL OF VIROLOGY Bd. 51, Nr. 3, September 1984, Seiten 650 - 655 SETH, P. ET AL. 'Role of a low-pH environment in adenovirus enhancement of the toxicity of a Pseudomonas exotoxin-Epidermal Growth Factor conjugate' *
MOLECULAR AND CELLULAR BIOLOGY Bd. 4, Nr. 8, August 1984, Seiten 1528 - 1533 SETH, P. ET AL. 'Evidence that the penton base of adenovirus is involved in potentiation of toxicity of Pseudomonas exotoxin conjugated to Epidermal Growth Factor' *
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA. Bd. 88, Nr. 19, 1. Oktober 1991, WASHINGTON US Seiten 8850 - 8854 CURIEL, D.T. ET AL. 'Adenovirus enhancement of transferrin-polylysine-mediated gene delivery' *
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA. Bd. 89, Nr. 13, 1. Juli 1992, WASHINGTON US Seiten 6094 - 6098 COTTEN, M. ET AL. 'High-efficiency receptor-mediated delivery of small and large (48 kilobase gene constructs using the endosome-disruption activity of defective or chemically inactivated adenovirus particles' *
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA. Bd. 89, Nr. 13, 1. Juli 1992, WASHINGTON US Seiten 6099 - 6103 WAGNER, E. ET AL. 'Coupling of adenovirus to transferrin-polylysine/DNA complexes greatly enhances receptor-mediated gene delivery and expression of transfected genes' *
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA. Bd. 89, Nr. 17, 1. September 1992, WASHINGTON US Seiten 7934 - 7938 WAGNER, E. ET AL. 'Influenza virus hemagglutinin HA-2 N-terminal fusogenic peptides augment gene transfer by transferrin-polylysine-DNA complexes: toward a synthetic virus-like gene-transfer vehicle' *

Cited By (159)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994006923A1 (en) * 1992-09-24 1994-03-31 The University Of Connecticut Modification of a virus to redirect infectivity and enhance targeted delivery of polynucleotides to cells
WO1994024299A1 (de) * 1993-04-08 1994-10-27 Boehringer Ingelheim International Gmbh Adenovirus für den transport von fremd-dna in höhere eukariotische zellen
US6020202A (en) * 1993-07-12 2000-02-01 Life Technologies, Inc. Composition and methods for transfecting eukaryotic cells
EP0654268A1 (en) * 1993-10-19 1995-05-24 BOEHRINGER INGELHEIM INTERNATIONAL GmbH A method to reverse the phenotype of transformed cells by the transcription factor IRF-1
US5658774A (en) * 1993-10-19 1997-08-19 Boehringer Ingelheim International Gmbh IRF-1 DNA expression inhibits growth of cells expressing c-myc or FosB
WO1995026411A3 (en) * 1994-03-25 1996-07-18 Uab Research Foundation Composition and methods for creating syngeneic recombinant virus-producing cells
WO1995026411A2 (en) * 1994-03-25 1995-10-05 The Uab Research Foundation Composition and methods for creating syngeneic recombinant virus-producing cells
WO1995033062A3 (de) * 1994-05-30 1996-07-18 Boehringer Ingelheim Int Verfahren zum einbringen von fremdmaterial in höhere eukaryotische zellen
WO1995033062A2 (de) * 1994-05-30 1995-12-07 Boehringer Ingelheim International Gmbh Verfahren zum einbringen von fremdmaterial in höhere eukaryotische zellen
US6284880B1 (en) 1994-05-30 2001-09-04 Boehringer Ingelheim International Gmbh Chicken embryo lethal orphan (CELO) virus GAM-1
US5965404A (en) * 1994-05-31 1999-10-12 Boehringer Ingelheim International Fmbh Method for introducing nucleic acids into higher eukaryotic cells
WO1996003517A1 (de) * 1994-07-26 1996-02-08 Boehringer Ingelheim International Gmbh Verfahren zur herstellung von viren und viralen vektoren
US6468981B1 (en) 1994-07-29 2002-10-22 Emory University Compositions and methods for targeting pharmaceutically active materials to cells containing androgen receptors
US5731190A (en) * 1994-09-08 1998-03-24 Genvec, Inc. Penton base protein and methods of using same
US5712136A (en) * 1994-09-08 1998-01-27 Genvec, Inc. Adenoviral-mediated cell targeting commanded by the adenovirus penton base protein
US6951755B2 (en) 1994-09-08 2005-10-04 Genvec, Inc. Vectors and methods for gene transfer
EP0789708A1 (en) * 1994-09-28 1997-08-20 Apollon, Inc. Multifunctional molecular complexes for gene transfer to cells
EP0789708A4 (en) * 1994-09-28 1999-12-29 Apollon Inc MULTIFUNCTIONAL MOLECULAR COMPLEXES FOR GENE TRANSFER TO CELLS
US6010871A (en) * 1994-09-29 2000-01-04 Ajinomoto Co., Inc. Modification of peptide and protein
US5770442A (en) * 1995-02-21 1998-06-23 Cornell Research Foundation, Inc. Chimeric adenoviral fiber protein and methods of using same
US6576456B2 (en) 1995-02-21 2003-06-10 Cornell Research Foundation, Inc. Chimeric adenovirus fiber protein
US6127525A (en) * 1995-02-21 2000-10-03 Cornell Research Foundation, Inc. Chimeric adenoviral coat protein and methods of using same
US6153435A (en) * 1995-02-21 2000-11-28 Cornell Research Foundation, Inc. Nucleic acid that encodes a chimeric adenoviral coat protein
WO1996029423A1 (en) * 1995-03-20 1996-09-26 Baylor College Of Medicine Compositions and methods for inducing infection by retroviral vectors outside of their host range
DE19510344C1 (de) * 1995-03-22 1996-11-07 Boehringer Ingelheim Int Verwendung einer Tumorvakzine
WO1996040958A1 (en) * 1995-06-07 1996-12-19 Baylor College Of Medicine Nucleic acid transporters for delivery of nucleic acids into a cell
US5736392A (en) * 1995-06-07 1998-04-07 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US6051429A (en) * 1995-06-07 2000-04-18 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US7105346B2 (en) 1995-06-15 2006-09-12 Crucell Holland B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
WO1997000965A2 (en) * 1995-06-23 1997-01-09 University Of Pittsburgh A lipidic vector for nucleic acid delivery
US5908777A (en) * 1995-06-23 1999-06-01 University Of Pittsburgh Lipidic vector for nucleic acid delivery
WO1997000965A3 (en) * 1995-06-23 1997-02-27 Univ Pittsburgh A lipidic vector for nucleic acid delivery
WO1997010851A2 (en) * 1995-09-07 1997-03-27 Opperbas Holding B.V. Nucleic acid delivery vehicle
WO1997010851A3 (en) * 1995-09-07 1997-06-05 Opperbas Holding Bv Nucleic acid delivery vehicle
US6057155A (en) * 1995-11-28 2000-05-02 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
US6649407B2 (en) 1995-11-28 2003-11-18 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
US5846782A (en) * 1995-11-28 1998-12-08 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
US6329190B1 (en) 1995-11-28 2001-12-11 Genvec, Inc. Targetting adenovirus with use of constrained peptide motifs
WO1997030170A1 (de) * 1996-02-15 1997-08-21 Boehringer Ingelheim International Gmbh Zusammensetzung für die transfektion höherer eukaryotischer zellen
WO1997033998A1 (en) * 1996-03-14 1997-09-18 The Immune Response Corporation Targeted delivery of genes encoding interferon
US6331525B1 (en) 1996-03-14 2001-12-18 The Immune Response Corporation Targeted delivery of genes encoding interferon
US6773709B2 (en) 1996-04-20 2004-08-10 Boehringer Ingelheim International Gmbh Chicken embryo lethal (CELO) virus
US6335016B1 (en) 1996-04-20 2002-01-01 Boehringer Ingelheim International Gmbh Chicken embryo lethal orphan (CELO) virus
DE19632532A1 (de) * 1996-08-13 1998-02-19 Boehringer Ingelheim Int Verfahren zur Herstellung von Säugetieren mit definierten genetischen Eigenschaften
EP0900849A1 (en) * 1996-10-31 1999-03-10 Shanghai Cancer Institute Receptor-mediated gene transfer system for targeting tumor gene therapy
EP0900849A4 (en) * 1996-10-31 2000-11-02 Shanghai Cancer Inst GENE TRANSFER SYSTEM THROUGH RECEPTORS FOR TARGETED GENE THERAPY OF TUMORS
EP2039768A1 (en) 1996-11-13 2009-03-25 Novartis Vaccines and Diagnostics, Inc. Mutant forms of Fas ligand and uses thereof
US6153596A (en) * 1996-12-18 2000-11-28 Emory University Polycationic oligomers
US6376248B1 (en) 1997-03-14 2002-04-23 Life Technologies, Inc. Peptide-enhanced transfections
EP2278006A2 (en) 1997-11-06 2011-01-26 Novartis Vaccines and Diagnostics S.r.l. Neisserial antigens
US7655245B2 (en) 1997-11-06 2010-02-02 Novartis Ag Neisserial antigens
US8293251B2 (en) 1997-11-06 2012-10-23 Novartis Ag Neisserial antigens
WO1999029349A1 (en) * 1997-12-04 1999-06-17 Boehringer Ingelheim International Gmbh Transfection particles
EP0945138A1 (en) * 1997-12-04 1999-09-29 Université Louis Pasteur de Strasbourg Transfection particles
EP2278011A2 (en) 1998-01-14 2011-01-26 Novartis Vaccines and Diagnostics S.r.l. Neisseria meningitidis antigens
EP2210945A2 (en) 1998-01-14 2010-07-28 Novartis Vaccines and Diagnostics S.r.l. Neisseria meningitidis antigens
US6599737B1 (en) 1998-04-30 2003-07-29 Cornell Research Foundation, Inc. Adenoviral vectors with tandem fiber proteins
EP2261349A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261357A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261352A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261347A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261355A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261338A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261350A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261353A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261348A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261341A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261339A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261345A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261351A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261346A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261343A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261344A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261340A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261354A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261356A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
EP2261342A2 (en) 1998-05-01 2010-12-15 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
US7749493B2 (en) 1998-07-08 2010-07-06 Crucell Holland B.V. Chimeric adenoviruses
WO2000011018A1 (en) * 1998-08-21 2000-03-02 Felix Frey Conjugates of dna interacting groups with steroid hormones for use as nucleic acid transfection agents
WO2000011019A1 (en) * 1998-08-21 2000-03-02 Felix Frey Conjugates of dna interacting groups with steroid hormones for use as nucleic acid transfection agent
US6455314B1 (en) 1998-09-11 2002-09-24 Genvec, Inc. Alternatively targeted adenovirus
US6914131B1 (en) 1998-10-09 2005-07-05 Chiron S.R.L. Neisserial antigens
EP1953229A2 (en) 1998-10-15 2008-08-06 Novartis Vaccines and Diagnostics, Inc. Metastatic breast and colon cancer regulated genes
US9358300B2 (en) 1998-11-12 2016-06-07 Life Technologies Corporation Transfection reagents
US6929946B1 (en) 1998-11-20 2005-08-16 Crucell Holland B.V. Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
EP1961813A2 (en) 1998-12-16 2008-08-27 Novartis Vaccines and Diagnostics, Inc. Human cyclin-dependent kinase (hPNQALRE)
EP2278007A1 (en) 1999-04-30 2011-01-26 Novartis Vaccines and Diagnostics S.r.l. Conserved neisserial antigens
EP2290083A1 (en) 1999-04-30 2011-03-02 Novartis Vaccines and Diagnostics S.r.l. Conserved neisserial antigens
US6913922B1 (en) 1999-05-18 2005-07-05 Crucell Holland B.V. Serotype of adenovirus and uses thereof
US7250293B2 (en) 1999-05-18 2007-07-31 Crucell Holland B.V. Complementing cell lines
US8221971B2 (en) 1999-05-18 2012-07-17 Crucell Holland B.V. Serotype of adenovirus and uses thereof
US7270811B2 (en) 1999-05-18 2007-09-18 Crucell Holland B.V. Serotype of adenovirus and uses thereof
EP2251424A1 (en) 1999-05-19 2010-11-17 Novartis Vaccines and Diagnostics S.r.l. Antigenic neisserial peptides
EP1935979A2 (en) 1999-07-14 2008-06-25 Novartis Vaccines and Diagnostics S.r.l. Antigenic meningococcal peptides
EP2275552A2 (en) 1999-10-29 2011-01-19 Novartis Vaccines and Diagnostics S.r.l. Neisserial antigenic peptides
EP2975127A1 (en) 1999-10-29 2016-01-20 GlaxoSmithKline Biologicals SA Neisserial antigenic peptides
EP2275554A2 (en) 1999-10-29 2011-01-19 Novartis Vaccines and Diagnostics S.r.l. Neisserial antigenic peptides
EP2275551A2 (en) 1999-10-29 2011-01-19 Novartis Vaccines and Diagnostics S.r.l. Neisserial antigenic peptides
EP2275553A2 (en) 1999-10-29 2011-01-19 Novartis Vaccines and Diagnostics S.r.l. Neisserial antigenic peptides
EP2289545A2 (en) 2000-01-17 2011-03-02 Novartis Vaccines and Diagnostics S.r.l. Supplemented OMV vaccine against meningococcus
EP2281570A2 (en) 2000-01-17 2011-02-09 Novartis Vaccines and Diagnostics S.r.l. Outer membrane vesicle (OMV) vaccine comprising n. meningitidis serogroup B outer membrane proteins
EP2281571A2 (en) 2000-01-17 2011-02-09 Novartis Vaccines and Diagnostics S.r.l. Outer membrane vesicle (omv) vaccine comprising n. meningitidids serogroup b outer membrane proteins
EP2275129A2 (en) 2000-01-17 2011-01-19 Novartis Vaccines and Diagnostics S.r.l. Outer membrane vesicle (OMV) vaccine comprising N. meningitidis serogroup B outer membrane proteins
US9267163B2 (en) 2000-02-28 2016-02-23 Glaxosmithkline Biologicals Sa Hybrid expression of neisserial proteins
EP1950297A2 (en) 2000-05-31 2008-07-30 Novartis Vaccines and Diagnostics, Inc. Compositions and methods for treating neoplastic disease using chemotherapy and radiation sensitizers
US9309520B2 (en) 2000-08-21 2016-04-12 Life Technologies Corporation Methods and compositions for synthesis of nucleic acid molecules using multiple recognition sites
US7235233B2 (en) 2000-09-26 2007-06-26 Crucell Holland B.V. Serotype 5 adenoviral vectors with chimeric fibers for gene delivery in skeletal muscle cells or myoblasts
EP2277895A1 (en) 2000-10-27 2011-01-26 Novartis Vaccines and Diagnostics S.r.l. Nucleic acids and proteins from streptococcus groups A & B
EP2284183A1 (en) 2000-10-27 2011-02-16 Novartis Vaccines and Diagnostics S.r.l. Nucleic acids and proteins from streptococcus groups A and B
EP2277894A1 (en) 2000-10-27 2011-01-26 Novartis Vaccines and Diagnostics S.r.l. Nucleic acids and proteins from streptococcus groups A & B
EP2284181A1 (en) 2000-10-27 2011-02-16 Novartis Vaccines and Diagnostics S.r.l. Nucleic acids and proteins from streptococcus groups A and B
EP2277896A1 (en) 2000-10-27 2011-01-26 Novartis Vaccines and Diagnostics S.r.l. Nucleic acids and proteins from streptococcus groups A & B
EP2896629A1 (en) 2000-10-27 2015-07-22 Novartis Vaccines and Diagnostics S.r.l. Nucleic acids and proteins from streptococcus group A & B
EP2284182A1 (en) 2000-10-27 2011-02-16 Novartis Vaccines and Diagnostics S.r.l. Nucleic acids and proteins from streptococcus groups A and B
US6974695B2 (en) 2000-11-15 2005-12-13 Crucell Holland B.V. Complementing cell lines
US7344883B2 (en) 2000-11-15 2008-03-18 Crucell Holland B.V. Complementing cell lines
US9228205B2 (en) 2000-11-15 2016-01-05 Crucell Holland B.V. Complementing cell lines
US8945884B2 (en) 2000-12-11 2015-02-03 Life Technologies Corporation Methods and compositions for synthesis of nucleic acid molecules using multiplerecognition sites
EP2278009A1 (en) 2001-03-27 2011-01-26 Novartis Vaccines and Diagnostics S.r.l. Streptococcus pneumoniae proteins and nucleic acids
EP2270176A1 (en) 2001-03-27 2011-01-05 Novartis Vaccines and Diagnostics S.r.l. Streptococcus pneumoniae proteins and nucleic acids
EP2278008A2 (en) 2001-03-27 2011-01-26 Novartis Vaccines and Diagnostics S.r.l. Streptococcus pneumoniae proteins and nucleic acids
EP2278010A1 (en) 2001-03-27 2011-01-26 Novartis Vaccines and Diagnostics S.r.l. Streptococcus pneumoniae proteins and nucleic acids
EP2298796A2 (en) 2001-03-27 2011-03-23 Novartis Vaccines and Diagnostics S.r.l. Staphylococcus aureus proteins and nucleic acids
EP2270177A1 (en) 2001-03-27 2011-01-05 Novartis Vaccines and Diagnostics S.r.l. Streptococcus pneumoniae proteins and nucleic acids
EP2314697A1 (en) 2001-03-27 2011-04-27 Novartis Vaccines and Diagnostics S.r.l. Streptococcus pneumoniae proteins and nucleic acids
EP2270175A1 (en) 2001-03-27 2011-01-05 Novartis Vaccines and Diagnostics S.r.l. Streptococcus pneumoniae proteins and nucleic acids
WO2003015825A3 (en) * 2001-08-16 2003-07-31 Photobiotics Ltd Conjugate
US7655753B2 (en) 2001-08-16 2010-02-02 Photobiotics Limited Conjugate
EP2335724A1 (en) 2001-12-12 2011-06-22 Novartis Vaccines and Diagnostics S.r.l. Immunisation against chlamydia trachomatis
EP2335723A1 (en) 2001-12-12 2011-06-22 Novartis Vaccines and Diagnostics S.r.l. Immunisation against chlamydia trachomatis
US7468181B2 (en) 2002-04-25 2008-12-23 Crucell Holland B.V. Means and methods for the production of adenovirus vectors
US7820440B2 (en) 2002-04-25 2010-10-26 Crucell Holland B.V. Means and methods for producing adenovirus vectors
EP2267005A1 (en) 2003-04-09 2010-12-29 Novartis Vaccines and Diagnostics S.r.l. ADP-ribosylating toxin from Listeria monocytogenes
US9090947B2 (en) 2003-10-23 2015-07-28 Kineta Two, Llc Detection of mutations in a gene associated with resistance to viral infection, OAS1
US9534252B2 (en) 2003-12-01 2017-01-03 Life Technologies Corporation Nucleic acid molecules containing recombination sites and methods of using the same
WO2005093064A1 (ja) 2004-03-29 2005-10-06 Galpharma Co., Ltd. 新規ガレクチン9改変体タンパク質及びその用途
WO2006042017A2 (en) 2004-10-06 2006-04-20 University Of Rochester Treatment of pulmonary hypertension using an agent that inhibits a tissue factor pathway
EP2110667A1 (en) 2005-01-20 2009-10-21 University Of Rochester Thoredoxin interacting protein (TXNIP) as regulator of vascular function
US9163222B2 (en) 2005-05-04 2015-10-20 Kineta Two, Llc Mutations in OAS1 genes
US8951768B2 (en) 2005-05-04 2015-02-10 Kineta Two, Llc Mutations in OAS1 genes
DE102005023993A1 (de) * 2005-05-20 2006-11-23 TransMIT Gesellschaft für Technologietransfer mbH Nicht virales Vektorsystem zum Transport von Nukleinsäure in die Lunge
WO2008020335A2 (en) 2006-06-09 2008-02-21 Novartis Ag Immunogenic compositions for streptococcus agalactiae
US8673859B2 (en) 2007-03-20 2014-03-18 New York University GM-CSF cosmeceutical compositions and methods of use thereof
WO2010107991A2 (en) 2009-03-18 2010-09-23 Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services Use of ixolaris, a tissue factor inhibitor, for the treatment and prevention of cancer
EP4218794A2 (en) 2009-06-10 2023-08-02 New York University Immunological targeting of pathological tau proteins
EP3329932A1 (en) 2009-06-10 2018-06-06 New York University Immunological targeting of pathological tau proteins
EP3553168A1 (en) 2010-11-12 2019-10-16 Georgetown University Immortalization of epithelial cells and methods of use
WO2012092341A1 (en) 2010-12-28 2012-07-05 University Of Rochester Methods of modifying insulin signaling using biliverdin reductase (bvr) and bvr derived peptides
WO2013036875A1 (en) 2011-09-07 2013-03-14 Mount Sinai School Of Medicine Ceramidase and cell differentiation
EP3741384A1 (en) 2011-09-07 2020-11-25 Mount Sinai School Of Medicine Ceramidase and cell differentiation
WO2014063155A1 (en) 2012-10-21 2014-04-24 University Of Rochester Thy1 (cd90) as a novel therapy to control adipose tissue accumulation
WO2014176259A1 (en) 2013-04-22 2014-10-30 Icahn School Of Medicine At Mount Sinai Mutations in pdgfrb and notch3 as causes of autosomal dominant infantile myofibromatosis
US11298376B2 (en) 2014-05-14 2022-04-12 Targimmune Therapeutics Ag Method of treating cancer
US10543232B2 (en) 2014-05-14 2020-01-28 Targimmune Therapeutics Ag Polyplex of double-stranded RNA and polymeric conjugate
US10792362B2 (en) 2014-07-15 2020-10-06 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
US10195280B2 (en) 2014-07-15 2019-02-05 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
US11872285B2 (en) 2014-07-15 2024-01-16 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
WO2017079442A1 (en) 2015-11-04 2017-05-11 Icahn School Of Medicine At Mount Sinai Methods of treating tumors and cancer, and identifying candidate subjects for such treatment
WO2017189730A1 (en) 2016-04-26 2017-11-02 Icahn School Of Medicine At Mount Sinai Treatment of hippo pathway mutant tumors and methods of identifying subjects as candidates for treatment

Also Published As

Publication number Publication date
HU211925A9 (en) 1996-01-29
IL103171A (en) 2003-04-10
ES2173083T3 (es) 2002-10-16
MX9205543A (es) 1993-05-01
EE03195B1 (sl) 1999-06-15
CZ293141B6 (cs) 2004-02-18
FI941474A (fi) 1994-03-30
BR9206559A (pt) 1994-11-08
NO316744B1 (no) 2004-04-26
HU9400898D0 (en) 1994-06-28
PL180304B1 (pl) 2001-01-31
NO941154L (no) 1994-03-29
HK1013104A1 (en) 1999-08-13
CZ74694A3 (en) 1995-05-17
ATE215990T1 (de) 2002-04-15
HU218846B (hu) 2000-12-28
CN1059705C (zh) 2000-12-20
NO941154D0 (no) 1994-03-29
SG44680A1 (en) 1997-12-19
US5547932A (en) 1996-08-20
AU671084B2 (en) 1996-08-15
HUT71312A (en) 1995-11-28
US6274322B1 (en) 2001-08-14
BG62740B1 (bg) 2000-06-30
US6022735A (en) 2000-02-08
CA2118816A1 (en) 1993-03-31
SK36894A3 (en) 1994-08-10
NZ244306A (en) 1995-07-26
DK0607206T3 (da) 2002-08-05
IL103171A0 (en) 1993-02-21
RO117861B1 (ro) 2002-08-30
BG98718A (bg) 1995-02-28
AU2652692A (en) 1993-05-03
CN1070946A (zh) 1993-04-14
SK281682B6 (sk) 2001-06-11
US6077663A (en) 2000-06-20
SI9200236B (sl) 2002-10-31
CA2118816C (en) 2003-06-17
EP0607206A1 (de) 1994-07-27
DE59209951D1 (de) 2002-05-16
EP0545016A1 (de) 1993-06-09
FI941474A0 (fi) 1994-03-30
JPH10506001A (ja) 1998-06-16
PT607206E (pt) 2002-07-31
EP0607206B1 (de) 2002-04-10
SI9200236A (en) 1993-03-31

Similar Documents

Publication Publication Date Title
EP0607206B1 (de) Zusammensetzung für das einbringen von nukleinsäure-komplexen in höhere eukaryotische zellen
US5981273A (en) Composition comprising an endosomolytic agent for introducing nucleic acid complexes into higher eucaryotic cells
EP0606280B1 (de) Neue konjugate zum einführen von nukleinsäure in höhere eukaryotische zellen
DE69534166T2 (de) Rekombinanter adenovirus und methoden zu dessen verwendung
WO1994021808A1 (de) Verfahren zur herstellung von krebsvakzinen
DE69838073T2 (de) Rekombinante nodavirus-zusammensetzungen und verfahren
DE69921435T2 (de) Verfahren zur Inaktivierung vom umhüllten Viren in einem Viruspräparat mit Adenoviren
CH684094A5 (de) Rekombinante Retroviren.
DE19910044A1 (de) Virale Partikel, die nach Infektion durch humanes Cytomegalovirus freigesetzt werden und ihre Verwendung als Impfstoff
DE4104186A1 (de) Neue, ueber endozytose in hoehere eukaryotische zellen aufnehmbare, nukleinsaeure enthaltende komplexe
DE4110409C2 (de) Neue Protein-Polykation-Konjugate
Ellison et al. Fusigenic liposome-mediated DNA transfer into cardiac myocytes
KR100241685B1 (ko) 핵산 복합체를 고등 진핵세포내로 도입시키기 위한 조성물
DE60036929T2 (de) Gentherapeutika
RU2138553C1 (ru) Трансфекционная композиция для высших эукариотных клеток, комплекс нуклеиновой кислоты, пригодный в качестве компонента трансфекционной композиции, конъюгат, пригодный в качестве компонента трансфекционной композиции, эндосомолитический пептид, пригодный в качестве компонента трансфекционной композиции
DE69728447T2 (de) Veränderte, kleine rna-viren
HRP920602A2 (hr) Sastav za uvođenje kompleksa nukleinskih kiselina u više eukariotične stanice
DE4110410C2 (de) Neue Protein-Polykation-Konjugate
DE4326821A1 (de) Verfahren zur Herstellung von Krebsvakzinen
AT399656B (de) Verfahren zur herstellung von krebsvakzinen
Huckett Gene transfer by receptor-mediated endocytosis: stable expression of NEO following insulin-directed entry into HepG2 cells.

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU BG BR CA CS FI HU JP KR NO PL RO

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
EX32 Extension under rule 32 effected after completion of technical preparation for international publication
LE32 Later election for international application filed prior to expiration of 19th month from priority date or according to rule 32.2 (b)
WWE Wipo information: entry into national phase

Ref document number: 2118816

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1019940700967

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 36894

Country of ref document: SK

Ref document number: 94-00499

Country of ref document: RO

WWE Wipo information: entry into national phase

Ref document number: PV1994-746

Country of ref document: CZ

Ref document number: 941474

Country of ref document: FI

WWE Wipo information: entry into national phase

Ref document number: 1992920580

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1992920580

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: PV1994-746

Country of ref document: CZ

WWG Wipo information: grant in national office

Ref document number: 1992920580

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: PV1994-746

Country of ref document: CZ